1
|
Song H, Li J, Yang H, Kong B, Xu Y, Li X, Li H. Enhancement of functional insulin-producing cell differentiation from embryonic stem cells through MST1-silencing. Diabetol Metab Syndr 2025; 17:93. [PMID: 40108649 PMCID: PMC11924671 DOI: 10.1186/s13098-025-01666-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 03/09/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Islet β-cell transplantation offers a promising treatment for repairing pancreatic damage in diabetes, with the transcription factor pancreatic duodenal homeobox-1 (PDX1) being crucial for β-cell function and insulin secretion. Mammalian threonine protein kinase (MST1) is recognized for its role in regulating PDX1 during cell apoptosis, yet its function in embryonic stem cell (ESC) differentiation into insulin-producing cells (IPCs) remain underexplored. This study investigated the effect of MST1-silencing on the differentiation of ESC into IPCs. METHODS ESCs were transfected utilizing a recombinant MST1-silencing lentiviral vector (shMST1). qRT-PCR, immunofluorescence, flow cytometry, western blot and ELISA assays were performed to examine function of IPCs in vitro. Furthermore, these IPCs were transplanted into type 1 diabetic mellitus (T1DM) rats. Measuring the changes in blood glucose concentration of animals before and after IPCs transplantation. Intraperitoneal glucose tolerance test (IPGT) was used to determine the regulatory effect of IPCs transplantation on blood glucose stimulation and immunohistochemistry was used to detect the expression of pancreatic Insulin protein in T1DM rats. RESULTS It was observed that IPCs from the shMST1 group exhibited notably improvement in insulin secretion and glucose responsiveness, suggesting MST1 suppression may enhance IPC maturity. The rats demonstrated significant normalization of blood sugar levels and increased insulin levels, akin to non-diabetic controls. This implies that MST1-silencing not only augments IPC function in vitro but also their therapeutic efficacy in vivo. CONCLUSIONS The findings indicate that targeting MST1 offers a novel approach for deriving functionally mature IPCs from ESCs, potentially advancing cell replacement therapies for diabetes. This research underscores the importance of developing IPCs with competent insulin secretion for diabetes treatment in vitro.
Collapse
Affiliation(s)
- Hui Song
- Basic Medical School of Ningxia Medical University, Yinchuan, 750004, China
- Institute of Endocrinology, Ningxia Medical University, Yinchuan, 750004, China
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, 750004, China
| | - Jiarui Li
- Basic Medical School of Ningxia Medical University, Yinchuan, 750004, China
- Institute of Endocrinology, Ningxia Medical University, Yinchuan, 750004, China
| | - Haohao Yang
- Basic Medical School of Ningxia Medical University, Yinchuan, 750004, China
- Institute of Endocrinology, Ningxia Medical University, Yinchuan, 750004, China
| | - Bin Kong
- Basic Medical School of Ningxia Medical University, Yinchuan, 750004, China
| | - Yu Xu
- Basic Medical School of Ningxia Medical University, Yinchuan, 750004, China
- Institute of Endocrinology, Ningxia Medical University, Yinchuan, 750004, China
| | - Xiong Li
- People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, 750004, China.
| | - Hui Li
- Basic Medical School of Ningxia Medical University, Yinchuan, 750004, China.
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, 750004, China.
| |
Collapse
|
2
|
Kang X, Ma L, Wen J, Gong W, Liu X, Hu Y, Feng Z, Jing Q, Cai Y, Li S, Cai X, Yuan K, Feng Y. Modeling of auditory neuropathy spectrum disorders associated with the TEME43 variant reveals impaired gap junction function of iPSC-derived glia-like support cells. Front Mol Neurosci 2025; 17:1457874. [PMID: 39834515 PMCID: PMC11743952 DOI: 10.3389/fnmol.2024.1457874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 11/22/2024] [Indexed: 01/22/2025] Open
Abstract
Auditory neuropathy spectrum disorder (ANSD) is an auditory dysfunction disorder characterized by impaired speech comprehension. Its etiology is complex and can be broadly categorized into genetic and non-genetic factors. TMEM43 mutation is identified as a causative factor in ANSD. While some studies have been conducted using animal models, its pathogenic mechanisms in humans remain unclear. TMEM43 is predominantly expressed in cochlear glia-like support cells (GLSs) and plays a vital role in gap junction intercellular communication. In this work, we utilized induced pluripotent stem cells from an ANSD patient carrying the TMEM43 gene mutation c.1114C>T (p.Arg372Ter) and directed their differentiation toward GLSs to investigate the effect of TMEM43 mutation on the function of gap junctions in cochlear GLSs in vitro. Reduced expression of genes associated with GLSs characteristics and reduced gap junction intercellular communication in TMEM43 mutant cell lines were observed compared to controls. Transcriptome analysis revealed that differentially expressed genes were significantly enriched in pathways related to cell proliferation, differentiation, extracellular space and adhesion. Furthermore, significant alterations were noted in the PI3K-Akt signaling pathway and the calcium signaling pathway, which could potentially influence gap junction function and contribute to hearing loss. In summary, our study based on patient-derived iPSCs sheds new light on the molecular mechanisms by which TMEM43 mutations may lead to ANSD. These mutations could result in developmental defects in GLSs and a diminished capacity for gap junction function, which may be implicated in the auditory deficits observed in ANSD patients. Our study explored the pathological effects of the TMEM43 mutation and its causal relationship with ANSD using a patient-derived iPSC-based GLSs model, providing a foundation for future mechanistic studies and potential drug screening efforts.
Collapse
Affiliation(s)
- Xiaoming Kang
- Department of Otorhinolaryngology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, University of South China, Changsha, China
| | - Lu Ma
- Institute of Otorhinolaryngology, Head and Neck Surgery, University of South China, Changsha, China
- MOE Key Lab of Rare Pediatric Diseases & Institute for Future Sciences, University of South China, Changsha, China
- Institute of Cytology and Genetics, Hengyang Medical School, University of South China, Hengyang, China
| | - Jie Wen
- Department of Otorhinolaryngology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, University of South China, Changsha, China
| | - Wei Gong
- Department of Otorhinolaryngology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, University of South China, Changsha, China
| | - Xianlin Liu
- Department of Otorhinolaryngology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, University of South China, Changsha, China
| | - Yihan Hu
- Department of Otorhinolaryngology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, University of South China, Changsha, China
| | - Zhili Feng
- Department of Otorhinolaryngology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, University of South China, Changsha, China
| | - Qiancheng Jing
- Department of Otorhinolaryngology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, University of South China, Changsha, China
| | - Yuexiang Cai
- Department of Otorhinolaryngology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, University of South China, Changsha, China
| | - Sijun Li
- Department of Otorhinolaryngology, Xiangya Hospital Central South University, Changsha, China
| | - Xinzhang Cai
- Department of Otorhinolaryngology, Xiangya Hospital Central South University, Changsha, China
| | - Kai Yuan
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Yong Feng
- Department of Otorhinolaryngology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, University of South China, Changsha, China
- MOE Key Lab of Rare Pediatric Diseases & Institute for Future Sciences, University of South China, Changsha, China
- Institute of Cytology and Genetics, Hengyang Medical School, University of South China, Hengyang, China
- Department of Otorhinolaryngology, Xiangya Hospital Central South University, Changsha, China
| |
Collapse
|
3
|
Manstein F, Halloin C, Zweigerdt R. Human Pluripotent Stem Cell Expansion in Stirred Tank Bioreactors. Methods Mol Biol 2025; 2924:59-71. [PMID: 40307635 DOI: 10.1007/978-1-0716-4530-7_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
This paper describes a detailed protocol for human pluripotent stem cells (hPSCs) cultivation as matrix-free cell-only aggregates in defined and xeno-free culture medium in stirred tank bioreactors (STBRs). Starting with a frozen stock pre-expanded on conventional culture dishes (2D), the ultimate process is performed in 150 mL culture scale in stirred tank bioreactors (3D) and is designed to produce up to 500 million pluripotent hPSC within 7 days. The culture strategy includes perfusion-based cell feeding facilitating process control, automation, and higher cell yields. Ultimately, this detailed protocol describes an important step for generating a defined starting cell population for directed lineage differentiation and subsequently fueling human cell-based assays and regenerative medicine approaches.
Collapse
Affiliation(s)
- Felix Manstein
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic-, Transplantation and Vascular Surgery, REBIRTH-Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Caroline Halloin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic-, Transplantation and Vascular Surgery, REBIRTH-Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Robert Zweigerdt
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic-, Transplantation and Vascular Surgery, REBIRTH-Cluster of Excellence, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
4
|
Garg P, Bähr M, Kügler S. Differentiation of Mature Dopaminergic Neurons from Human Induced Pluripotent Stem Cells. Methods Mol Biol 2025; 2910:53-68. [PMID: 40220093 DOI: 10.1007/978-1-0716-4446-1_4] [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] [Indexed: 04/14/2025]
Abstract
Reprogramming of somatic cells like blood cells and fibroblasts to obtain human induced pluripotent stem cells (hiPSCs) has become a state-of-the-art tool to study human diseases. The self-renewable hiPSCs offer ease of genetic modifications and can be differentiated into any cell type of the human body ranging from hepatocytes, cardiac myocytes, and subtypes of neurons. Dopaminergic (DA) neurons are one such neuronal subtype that is largely present in the midbrain region of the human brain and controls several functions like voluntary movement, reward, addiction, and stress. Loss of DA neurons is associated with one of the most common neurological disorders, Parkinson's disease (PD). Here, we describe a small molecule-directed approach for the generation of functionally mature dopaminergic neurons through the differentiation of hiPSCs. Differentiated DA neurons can be used to study their role in (patho)physiology.
Collapse
Affiliation(s)
- Pretty Garg
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany.
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany.
| | - Mathias Bähr
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Sebastian Kügler
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
5
|
Romito E, Battistella I, Plakhova V, Paplekaj A, Forastieri C, Toffolo E, Musio C, Conti L, Battaglioli E, Rusconi F. A comprehensive protocol for efficient differentiation of human NPCs into electrically competent neurons. J Neurosci Methods 2024; 410:110225. [PMID: 39053772 DOI: 10.1016/j.jneumeth.2024.110225] [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: 04/08/2024] [Revised: 05/29/2024] [Accepted: 07/19/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND The study of neurons is fundamental to unraveling the complexities of the nervous system. Primary neuronal cultures from rodents have long been a cornerstone of experimental studies, yet limitations related to their non-human nature and ethical concerns have prompted the development of alternatives. In recent years, the derivation of neurons from human-induced pluripotent stem cells (hiPSCs) has emerged as a powerful option, offering a scalable source of cells for diverse applications. Neural progenitor cells (NPCs) derived from hiPSCs can be efficiently differentiated into functional neurons, providing a platform to study human neural physiology and pathology in vitro. However, challenges persist in achieving consistent and reproducible outcomes across experimental settings. COMPARISON WITH EXISTING METHODS Our aim is to provide a step-by-step methodological protocol, augmenting existing procedures with additional instructions and parameters, to guide researchers in achieving reproducible results. METHODS AND RESULTS We outline procedures for the differentiation of hiPSC-derived NPCs into electrically competent neurons, encompassing initial cell density, morphology, maintenance, and differentiation. We also describe the analysis of specific markers for assessing neuronal phenotype, along with electrophysiological analysis to evaluate biophysical properties of neuronal excitability. Additionally, we conduct a comparative analysis of three different chemical methods-KCl, N-methyl-D-aspartate (NMDA), and bicuculline-to induce neuronal depolarization and assess their effects on the induction of both fast and slow post-translational, transcriptional, and post-transcriptional responses. CONCLUSION Our protocol provides clear instructions for generating reliable human neuronal cultures with defined electrophysiological properties to investigate neuronal differentiation and model diseases in vitro.
Collapse
Affiliation(s)
- Elena Romito
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Via Fratelli Cervi, 93, Segrate, Milan 20054, Italy
| | - Ingrid Battistella
- Department of Cellular, Computational and Integrative Biology - CIBIO, Università degli Studi di Trento, Via Sommarive, 9, Trento 38123, Italy
| | - Vera Plakhova
- Institute of Biophysics (IBF), National Research Council (CNR), Trento Unit, & LabSSAH, Bruno Kessler Foundation (FBK), Via Sommarive, 18, Trento 38123, Italy
| | - Arteda Paplekaj
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Via Fratelli Cervi, 93, Segrate, Milan 20054, Italy
| | - Chiara Forastieri
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Via Fratelli Cervi, 93, Segrate, Milan 20054, Italy
| | - Emanuela Toffolo
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Via Fratelli Cervi, 93, Segrate, Milan 20054, Italy
| | - Carlo Musio
- Institute of Biophysics (IBF), National Research Council (CNR), Trento Unit, & LabSSAH, Bruno Kessler Foundation (FBK), Via Sommarive, 18, Trento 38123, Italy
| | - Luciano Conti
- Department of Cellular, Computational and Integrative Biology - CIBIO, Università degli Studi di Trento, Via Sommarive, 9, Trento 38123, Italy.
| | - Elena Battaglioli
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Via Fratelli Cervi, 93, Segrate, Milan 20054, Italy
| | - Francesco Rusconi
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Via Fratelli Cervi, 93, Segrate, Milan 20054, Italy.
| |
Collapse
|
6
|
Munezane H, Imamura K, Fujimoto N, Hotta A, Yukitake H, Inoue H. Elimination of the extra chromosome of Dup15q syndrome iPSCs for cellular and molecular investigation. Eur J Cell Biol 2024; 103:151446. [PMID: 39059105 DOI: 10.1016/j.ejcb.2024.151446] [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: 04/17/2024] [Revised: 06/23/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Chromosome 15q11.2-13.1 duplication (Dup15q) syndrome is one of the most common autism spectrum disorders (ASDs) associated with copy number variants (CNVs). For the analysis of CNV-relevant pathological cellular phenotypes, a CNV-corrected isogenic cell line is useful for excluding the influence of genetic background. Here, we devised a strategy to remove the isodicentric chromosome 15 by inserting a puro-ΔTK selection cassette into the extra chromosome using the CRISPR-Cas9 system, followed by a subsequent two-step drug selection. A series of assays, including qPCR-based copy number analysis and karyotype analysis, confirmed the elimination of the extra chromosome. Furthermore, cerebral organoids were generated from the parental Dup15q iPSCs and their isogenic iPSCs. scRNA-seq analysis revealed the alteration of expression levels in ion-channel-related genes and synapse-related genes in glutamatergic and GABAergic neurons in Dup15q organoids, respectively. The established isogenic cell line is a valuable resource for unraveling cellular and molecular alterations associated with Dup15q syndrome.
Collapse
Affiliation(s)
- Haruka Munezane
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan; Takeda-CiRA (T-CiRA) Joint Program, 2-26-1, Muraoka-Higashi, Fujisawa 251-8555, Japan
| | - Keiko Imamura
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan; Takeda-CiRA (T-CiRA) Joint Program, 2-26-1, Muraoka-Higashi, Fujisawa 251-8555, Japan; iPSC-based Drug discovery and Development Team, RIKEN BioResource Research Center, 1-7 Hikaridai, Seika-cho, Soraku-gun, Kyoto 619-0237, Japan; Medical-Risk Avoidance based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Naoko Fujimoto
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan; Takeda-CiRA (T-CiRA) Joint Program, 2-26-1, Muraoka-Higashi, Fujisawa 251-8555, Japan
| | - Akitsu Hotta
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan; Takeda-CiRA (T-CiRA) Joint Program, 2-26-1, Muraoka-Higashi, Fujisawa 251-8555, Japan
| | - Hiroshi Yukitake
- Takeda-CiRA (T-CiRA) Joint Program, 2-26-1, Muraoka-Higashi, Fujisawa 251-8555, Japan; Global Advanced Platform, Takeda Pharmaceutical Company Limited, 2-26-1, Muraoka-Higashi, Fujisawa 251-8555, Japan
| | - Haruhisa Inoue
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan; Takeda-CiRA (T-CiRA) Joint Program, 2-26-1, Muraoka-Higashi, Fujisawa 251-8555, Japan; iPSC-based Drug discovery and Development Team, RIKEN BioResource Research Center, 1-7 Hikaridai, Seika-cho, Soraku-gun, Kyoto 619-0237, Japan; Medical-Risk Avoidance based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan.
| |
Collapse
|
7
|
Kriedemann N, Manstein F, Hernandez-Bautista CA, Ullmann K, Triebert W, Franke A, Mertens M, Stein ICAP, Leffler A, Witte M, Askurava T, Fricke V, Gruh I, Piep B, Kowalski K, Kraft T, Zweigerdt R. Protein-free media for cardiac differentiation of hPSCs in 2000 mL suspension culture. Stem Cell Res Ther 2024; 15:213. [PMID: 39020441 PMCID: PMC11256493 DOI: 10.1186/s13287-024-03826-w] [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: 05/16/2024] [Accepted: 07/01/2024] [Indexed: 07/19/2024] Open
Abstract
BACKGROUND Commonly used media for the differentiation of human pluripotent stem cells into cardiomyocytes (hPSC-CMs) contain high concentrations of proteins, in particular albumin, which is prone to quality variations and presents a substantial cost factor, hampering the clinical translation of in vitro-generated cardiomyocytes for heart repair. To overcome these limitations, we have developed chemically defined, entirely protein-free media based on RPMI, supplemented with L-ascorbic acid 2-phosphate (AA-2P) and either the non-ionic surfactant Pluronic F-68 or a specific polyvinyl alcohol (PVA). METHODS AND RESULTS Both media compositions enable the efficient, directed differentiation of embryonic and induced hPSCs, matching the cell yields and cardiomyocyte purity ranging from 85 to 99% achieved with the widely used protein-based CDM3 medium. The protein-free differentiation approach was readily up-scaled to a 2000 mL process scale in a fully controlled stirred tank bioreactor in suspension culture, producing > 1.3 × 109 cardiomyocytes in a single process run. Transcriptome analysis, flow cytometry, electrophysiology, and contractile force measurements revealed that the mass-produced cardiomyocytes differentiated in protein-free medium exhibit the expected ventricular-like properties equivalent to the well-established characteristics of CDM3-control cells. CONCLUSIONS This study promotes the robustness and upscaling of the cardiomyogenic differentiation process, substantially reduces media costs, and provides an important step toward the clinical translation of hPSC-CMs for heart regeneration.
Collapse
Affiliation(s)
- Nils Kriedemann
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO)Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG)REBIRTH - Research Center for Translational Regenerative Medicine, Hannover Medical School (MHH), Carl Neuberg-Str. 1, 30625, Hannover, Germany.
| | - Felix Manstein
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO)Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG)REBIRTH - Research Center for Translational Regenerative Medicine, Hannover Medical School (MHH), Carl Neuberg-Str. 1, 30625, Hannover, Germany
- Evotec SE, Hamburg, Germany
| | - Carlos A Hernandez-Bautista
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO)Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG)REBIRTH - Research Center for Translational Regenerative Medicine, Hannover Medical School (MHH), Carl Neuberg-Str. 1, 30625, Hannover, Germany
| | - Kevin Ullmann
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO)Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG)REBIRTH - Research Center for Translational Regenerative Medicine, Hannover Medical School (MHH), Carl Neuberg-Str. 1, 30625, Hannover, Germany
| | - Wiebke Triebert
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO)Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG)REBIRTH - Research Center for Translational Regenerative Medicine, Hannover Medical School (MHH), Carl Neuberg-Str. 1, 30625, Hannover, Germany
- Evotec SE, Hamburg, Germany
| | - Annika Franke
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO)Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG)REBIRTH - Research Center for Translational Regenerative Medicine, Hannover Medical School (MHH), Carl Neuberg-Str. 1, 30625, Hannover, Germany
| | - Mira Mertens
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO)Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG)REBIRTH - Research Center for Translational Regenerative Medicine, Hannover Medical School (MHH), Carl Neuberg-Str. 1, 30625, Hannover, Germany
| | | | - Andreas Leffler
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School (MHH), Hannover, Germany
| | - Merlin Witte
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO)Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG)REBIRTH - Research Center for Translational Regenerative Medicine, Hannover Medical School (MHH), Carl Neuberg-Str. 1, 30625, Hannover, Germany
| | - Tamari Askurava
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO)Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG)REBIRTH - Research Center for Translational Regenerative Medicine, Hannover Medical School (MHH), Carl Neuberg-Str. 1, 30625, Hannover, Germany
| | - Veronika Fricke
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO)Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG)REBIRTH - Research Center for Translational Regenerative Medicine, Hannover Medical School (MHH), Carl Neuberg-Str. 1, 30625, Hannover, Germany
| | - Ina Gruh
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO)Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG)REBIRTH - Research Center for Translational Regenerative Medicine, Hannover Medical School (MHH), Carl Neuberg-Str. 1, 30625, Hannover, Germany
| | - Birgit Piep
- Institute of Molecular and Cell Physiology, Hannover Medical School (MHH), Hannover, Germany
| | - Kathrin Kowalski
- Institute of Molecular and Cell Physiology, Hannover Medical School (MHH), Hannover, Germany
| | - Theresia Kraft
- Institute of Molecular and Cell Physiology, Hannover Medical School (MHH), Hannover, Germany
| | - Robert Zweigerdt
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO)Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG)REBIRTH - Research Center for Translational Regenerative Medicine, Hannover Medical School (MHH), Carl Neuberg-Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
8
|
Yin X, Li Q, Shu Y, Wang H, Thomas B, Maxwell JT, Zhang Y. Exploiting urine-derived induced pluripotent stem cells for advancing precision medicine in cell therapy, disease modeling, and drug testing. J Biomed Sci 2024; 31:47. [PMID: 38724973 PMCID: PMC11084032 DOI: 10.1186/s12929-024-01035-4] [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: 01/26/2024] [Accepted: 04/30/2024] [Indexed: 05/12/2024] Open
Abstract
The field of regenerative medicine has witnessed remarkable advancements with the emergence of induced pluripotent stem cells (iPSCs) derived from a variety of sources. Among these, urine-derived induced pluripotent stem cells (u-iPSCs) have garnered substantial attention due to their non-invasive and patient-friendly acquisition method. This review manuscript delves into the potential and application of u-iPSCs in advancing precision medicine, particularly in the realms of drug testing, disease modeling, and cell therapy. U-iPSCs are generated through the reprogramming of somatic cells found in urine samples, offering a unique and renewable source of patient-specific pluripotent cells. Their utility in drug testing has revolutionized the pharmaceutical industry by providing personalized platforms for drug screening, toxicity assessment, and efficacy evaluation. The availability of u-iPSCs with diverse genetic backgrounds facilitates the development of tailored therapeutic approaches, minimizing adverse effects and optimizing treatment outcomes. Furthermore, u-iPSCs have demonstrated remarkable efficacy in disease modeling, allowing researchers to recapitulate patient-specific pathologies in vitro. This not only enhances our understanding of disease mechanisms but also serves as a valuable tool for drug discovery and development. In addition, u-iPSC-based disease models offer a platform for studying rare and genetically complex diseases, often underserved by traditional research methods. The versatility of u-iPSCs extends to cell therapy applications, where they hold immense promise for regenerative medicine. Their potential to differentiate into various cell types, including neurons, cardiomyocytes, and hepatocytes, enables the development of patient-specific cell replacement therapies. This personalized approach can revolutionize the treatment of degenerative diseases, organ failure, and tissue damage by minimizing immune rejection and optimizing therapeutic outcomes. However, several challenges and considerations, such as standardization of reprogramming protocols, genomic stability, and scalability, must be addressed to fully exploit u-iPSCs' potential in precision medicine. In conclusion, this review underscores the transformative impact of u-iPSCs on advancing precision medicine and highlights the future prospects and challenges in harnessing this innovative technology for improved healthcare outcomes.
Collapse
Affiliation(s)
- Xiya Yin
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yan Shu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Baltimore, MD, USA
| | - Hongbing Wang
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Baltimore, MD, USA
| | - Biju Thomas
- Keck School of Medicine, Roski Eye Institute, University of Southern California, Los Angeles, CA, 90033, USA
| | - Joshua T Maxwell
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Yuanyuan Zhang
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA.
| |
Collapse
|
9
|
Triebelhorn J, Cardon I, Kuffner K, Bader S, Jahner T, Meindl K, Rothhammer-Hampl T, Riemenschneider MJ, Drexler K, Berneburg M, Nothdurfter C, Manook A, Brochhausen C, Baghai TC, Hilbert S, Rupprecht R, Milenkovic VM, Wetzel CH. Induced neural progenitor cells and iPS-neurons from major depressive disorder patients show altered bioenergetics and electrophysiological properties. Mol Psychiatry 2024; 29:1217-1227. [PMID: 35732695 PMCID: PMC11189806 DOI: 10.1038/s41380-022-01660-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 05/30/2022] [Accepted: 06/07/2022] [Indexed: 11/09/2022]
Abstract
The molecular pathomechanisms of major depressive disorder (MDD) are still not completely understood. Here, we follow the hypothesis, that mitochondria dysfunction which is inevitably associated with bioenergetic disbalance is a risk factor that contributes to the susceptibility of an individual to develop MDD. Thus, we investigated molecular mechanisms related to mitochondrial function in induced neuronal progenitor cells (NPCs) which were reprogrammed from fibroblasts of eight MDD patients and eight non-depressed controls. We found significantly lower maximal respiration rates, altered cytosolic basal calcium levels, and smaller soma size in NPCs derived from MDD patients. These findings are partially consistent with our earlier observations in MDD patient-derived fibroblasts. Furthermore, we differentiated MDD and control NPCs into iPS-neurons and analyzed their passive biophysical and active electrophysiological properties to investigate whether neuronal function can be related to altered mitochondrial activity and bioenergetics. Interestingly, MDD patient-derived iPS-neurons showed significantly lower membrane capacitance, a less hyperpolarized membrane potential, increased Na+ current density and increased spontaneous electrical activity. Our findings indicate that functional differences evident in fibroblasts derived from MDD patients are partially present after reprogramming to induced-NPCs, could relate to altered function of iPS-neurons and thus might be associated with the aetiology of major depressive disorder.
Collapse
Affiliation(s)
- Julian Triebelhorn
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053, Regensburg, Germany
| | - Iseline Cardon
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053, Regensburg, Germany
| | - Kerstin Kuffner
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053, Regensburg, Germany
| | - Stefanie Bader
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053, Regensburg, Germany
| | - Tatjana Jahner
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053, Regensburg, Germany
| | - Katrin Meindl
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053, Regensburg, Germany
| | - Tanja Rothhammer-Hampl
- Department of Neuropathology, Regensburg University Hospital, 93053, Regensburg, Germany
| | | | - Konstantin Drexler
- Department of Dermatology, Regensburg University Hospital, 93053, Regensburg, Germany
| | - Mark Berneburg
- Department of Dermatology, Regensburg University Hospital, 93053, Regensburg, Germany
| | - Caroline Nothdurfter
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053, Regensburg, Germany
| | - André Manook
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053, Regensburg, Germany
| | - Christoph Brochhausen
- Institute of Pathology, University of Regensburg, 93053, Regensburg, Germany
- Central Biobank of the University of Regensburg and the Regensburg University Hospital, 93053, Regensburg, Germany
| | - Thomas C Baghai
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053, Regensburg, Germany
| | - Sven Hilbert
- Institute of Educational Research, Faculty of Human Sciences, University of Regensburg, 93053, Regensburg, Germany
| | - Rainer Rupprecht
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053, Regensburg, Germany
| | - Vladimir M Milenkovic
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053, Regensburg, Germany
| | - Christian H Wetzel
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053, Regensburg, Germany.
| |
Collapse
|
10
|
Lewis J, Yaseen B, Saraf A. Novel 2D/3D Hybrid Organoid System for High-Throughput Drug Screening in iPSC Cardiomyocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.29.591754. [PMID: 38746465 PMCID: PMC11092641 DOI: 10.1101/2024.04.29.591754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Human induced pluripotent stem cell cardiomyocytes (hiPSC-CMs) allow for high-throughput evaluation of cardiomyocyte (CM) physiology in health and disease. While multimodality testing provides a large breadth of information related to electrophysiology, contractility, and intracellular signaling in small populations of iPSC-CMs, current technologies for analyzing these parameters are expensive and resource-intensive. We sought to design a 2D/3D hybrid organoid system and harness optical imaging techniques to assess electromechanical properties, calcium dynamics, and signal propagation across CMs in a high-throughput manner. We validated our methods using a doxorubicin-based system, as the drug has well-characterized cardiotoxic, pro-arrhythmic effects. hiPSCs were differentiated into CMs, assembled into organoids, and thereafter treated with doxorubicin. The organoids were then replated to form a hybrid 2D/3D iPSC-CM construct where the 3D cardiac organoids acted as the source of electromechanical activity which propagated outwards into a 2D iPSC-CM sheet. The organoid recapitulated cardiac structure and connectivity, while 2D CMs facilitated analysis at an individual cellular level which recreated numerous doxorubicin-induced electrophysiologic and propagation abnormalities. Thus, we have developed a novel 2D/3D hybrid organoid model that employs an integrated optical analysis platform to provide a reliable high-throughput method for studying cardiotoxicity, providing valuable data on calcium, contractility, and signal propagation.
Collapse
|
11
|
Ullmann K, Manstein F, Triebert W, Kriedemann N, Franke A, Teske J, Mertens M, Lupanow V, Göhring G, Haase A, Martin U, Zweigerdt R. Matrix-free human pluripotent stem cell manufacturing by seed train approach and intermediate cryopreservation. Stem Cell Res Ther 2024; 15:89. [PMID: 38528578 DOI: 10.1186/s13287-024-03699-z] [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: 01/10/2024] [Accepted: 03/17/2024] [Indexed: 03/27/2024] Open
Abstract
BACKGROUND Human pluripotent stem cells (hPSCs) have an enormous therapeutic potential, but large quantities of cells will need to be supplied by reliable, economically viable production processes. The suspension culture (three-dimensional; 3D) of hPSCs in stirred tank bioreactors (STBRs) has enormous potential for fuelling these cell demands. In this study, the efficient long-term matrix-free suspension culture of hPSC aggregates is shown. METHODS AND RESULTS STBR-controlled, chemical aggregate dissociation and optimized passage duration of 3 or 4 days promotes exponential hPSC proliferation, process efficiency and upscaling by a seed train approach. Intermediate high-density cryopreservation of suspension-derived hPSCs followed by direct STBR inoculation enabled complete omission of matrix-dependent 2D (two-dimensional) culture. Optimized 3D cultivation over 8 passages (32 days) cumulatively yielded ≈4.7 × 1015 cells, while maintaining hPSCs' pluripotency, differentiation potential and karyotype stability. Gene expression profiling reveals novel insights into the adaption of hPSCs to continuous 3D culture compared to conventional 2D controls. CONCLUSIONS Together, an entirely matrix-free, highly efficient, flexible and automation-friendly hPSC expansion strategy is demonstrated, facilitating the development of good manufacturing practice-compliant closed-system manufacturing in large scale.
Collapse
Affiliation(s)
- Kevin Ullmann
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic Transplantation and Vascular Surgery (HTTG), Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
- REBIRTH Research Center for Translational and Regenerative Medicine, Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| | - Felix Manstein
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic Transplantation and Vascular Surgery (HTTG), Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- REBIRTH Research Center for Translational and Regenerative Medicine, Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Wiebke Triebert
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic Transplantation and Vascular Surgery (HTTG), Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- REBIRTH Research Center for Translational and Regenerative Medicine, Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Nils Kriedemann
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic Transplantation and Vascular Surgery (HTTG), Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- REBIRTH Research Center for Translational and Regenerative Medicine, Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Annika Franke
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic Transplantation and Vascular Surgery (HTTG), Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- REBIRTH Research Center for Translational and Regenerative Medicine, Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Jana Teske
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic Transplantation and Vascular Surgery (HTTG), Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- REBIRTH Research Center for Translational and Regenerative Medicine, Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Mira Mertens
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic Transplantation and Vascular Surgery (HTTG), Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- REBIRTH Research Center for Translational and Regenerative Medicine, Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Victoria Lupanow
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic Transplantation and Vascular Surgery (HTTG), Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- REBIRTH Research Center for Translational and Regenerative Medicine, Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Gudrun Göhring
- Department of Human Genetics, Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Alexandra Haase
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic Transplantation and Vascular Surgery (HTTG), Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- REBIRTH Research Center for Translational and Regenerative Medicine, Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Ulrich Martin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic Transplantation and Vascular Surgery (HTTG), Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- REBIRTH Research Center for Translational and Regenerative Medicine, Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Robert Zweigerdt
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic Transplantation and Vascular Surgery (HTTG), Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
- REBIRTH Research Center for Translational and Regenerative Medicine, Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
12
|
Agriesti F, Cela O, Capitanio N. "Time Is out of Joint" in Pluripotent Stem Cells: How and Why. Int J Mol Sci 2024; 25:2063. [PMID: 38396740 PMCID: PMC10889767 DOI: 10.3390/ijms25042063] [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: 01/10/2024] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
The circadian rhythm is necessary for the homeostasis and health of living organisms. Molecular clocks interconnected by transcription/translation feedback loops exist in most cells of the body. A puzzling exemption to this, otherwise, general biological hallmark is given by the cell physiology of pluripotent stem cells (PSCs) that lack circadian oscillations gradually acquired following their in vivo programmed differentiation. This process can be nicely phenocopied following in vitro commitment and reversed during the reprogramming of somatic cells to induce PSCs. The current understanding of how and why pluripotency is "time-uncoupled" is largely incomplete. A complex picture is emerging where the circadian core clockwork is negatively regulated in PSCs at the post-transcriptional/translational, epigenetic, and other-clock-interaction levels. Moreover, non-canonical functions of circadian core-work components in the balance between pluripotency identity and metabolic-driven cell reprogramming are emerging. This review selects and discusses results of relevant recent investigations providing major insights into this context.
Collapse
Affiliation(s)
- Francesca Agriesti
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (O.C.); (N.C.)
| | | | | |
Collapse
|
13
|
Khandani B, Movahedin M. Learning Towards Maturation of Defined Feeder-free Pluripotency Culture Systems: Lessons from Conventional Feeder-based Systems. Stem Cell Rev Rep 2024; 20:484-494. [PMID: 38079087 DOI: 10.1007/s12015-023-10662-7] [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] [Accepted: 12/01/2023] [Indexed: 02/03/2024]
Abstract
Pluripotent stem cells (PSCs) are widely recognized as one of the most promising types of stem cells for applications in regenerative medicine, tissue engineering, disease modeling, and drug screening. This is due to their unique ability to differentiate into cells from all three germ layers and their capacity for indefinite self-renewal. Initially, PSCs were cultured using animal feeder cells, but these systems presented several limitations, particularly in terms of Good Manufacturing Practices (GMP) regulations. As a result, feeder-free systems were introduced as a safer alternative. However, the precise mechanisms by which feeder cells support pluripotency are not fully understood. More importantly, it has been observed that some aspects of the need for feeder cells like the optimal density and cell type can vary depending on conditions such as the developmental stage of the PSCs, phases of the culture protocol, the method used in culture for induction of pluripotency, and intrinsic variability of PSCs. Thus, gaining a better understanding of the divergent roles and necessity of feeder cells in various conditions would lead to the development of condition-specific defined feeder-free systems that resolve the failure of current feeder-free systems in some conditions. Therefore, this review aims to explore considerable feeder-related issues that can lead to the development of condition-specific feeder-free systems.
Collapse
Affiliation(s)
- Bardia Khandani
- Department of Stem Cells Technology and Tissue Regeneration, Faculty of Interdisciplinary Science and Technology, Tarbiat Modares University, Tehran, Iran
| | - Mansoureh Movahedin
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Jalal Ale Ahmad Highway, Tehran, 14115111, Iran.
| |
Collapse
|
14
|
Wen J, Song J, Chen J, Feng Z, Jing Q, Gong W, Kang X, Mei L, He C, Ma L, Feng Y. Modeling of pigmentation disorders associated with MITF mutation in Waardenburg syndrome revealed an impaired melanogenesis pathway in iPS-derived melanocytes. Pigment Cell Melanoma Res 2024; 37:21-35. [PMID: 37559350 DOI: 10.1111/pcmr.13118] [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: 09/20/2022] [Revised: 06/18/2023] [Accepted: 07/26/2023] [Indexed: 08/11/2023]
Abstract
Waardenburg Syndrome (WS) is a rare genetic disorder that leads to congenital hearing loss and pigmentation defects. Microphthalmia-associated transcription factor (MITF) is one of its significant pathogenic genes. Despite the comprehensive investigation in animal models, the pathogenic mechanism is still poorly described in humans due to difficulties accessing embryonic tissues. In this work, we used induced pluripotent stem cells derived from a WS patient carrying a heterozygous mutation in the MITF gene c.626A>T (p.His209Leu), and differentiated toward melanocyte lineage, which is the most affected cell type involved in WS. Compared with the wild-type cell line, the MITFmut cell line showed a reduced expression of the characteristic melanocyte-related genes and a lesser proportion of mature, fully pigmented melanosomes. The transcriptome analysis also revealed widespread gene expression changes at the melanocyte stage in the MITFmut cell line. The differentially expressed genes were enriched in melanogenesis and cell proliferation-related pathways. Interestingly, ion transport-related genes also showed a significant difference in MITFmut -induced melanocytes, indicating that the MITF mutant may lead to the dysfunction of potassium channels and transporters produced by intermediate cells in the cochlea, further causing the associated phenotype of deafness. Altogether, our study provides valuable insights into how MITF mutation affects WS patients, which might result in defective melanocyte development and the related phenotype based on the patient-derived iPSC model.
Collapse
Affiliation(s)
- Jie Wen
- Department of Otorhinolaryngology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, University of South China, Changsha, China
| | - Jian Song
- Department of Otorhinolaryngology, Xiangya Hospital Central South University, Changsha, China
- Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, China
| | - Jiale Chen
- Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Zhili Feng
- Department of Otorhinolaryngology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, University of South China, Changsha, China
| | - Qiancheng Jing
- Department of Otorhinolaryngology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, University of South China, Changsha, China
| | - Wei Gong
- Department of Otorhinolaryngology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, University of South China, Changsha, China
| | - Xiaoming Kang
- Department of Otorhinolaryngology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, University of South China, Changsha, China
| | - Lingyun Mei
- Department of Otorhinolaryngology, Xiangya Hospital Central South University, Changsha, China
- Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, China
| | - Chufeng He
- Department of Otorhinolaryngology, Xiangya Hospital Central South University, Changsha, China
- Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, China
| | - Lu Ma
- Institute of Otorhinolaryngology, Head and Neck Surgery, University of South China, Changsha, China
- The Hengyang Key Laboratory of Cellular Stress Biology, Institute of Cytology and Genetics, Hengyang Medical School, University of South China, Hengyang, China
| | - Yong Feng
- Department of Otorhinolaryngology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Institute of Otorhinolaryngology, Head and Neck Surgery, University of South China, Changsha, China
- Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, China
- Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
15
|
Hashizume T, Ying BW. Challenges in developing cell culture media using machine learning. Biotechnol Adv 2024; 70:108293. [PMID: 37984683 DOI: 10.1016/j.biotechadv.2023.108293] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 10/17/2023] [Accepted: 11/14/2023] [Indexed: 11/22/2023]
Abstract
Microbial and mammalian cells are widely used in the food, pharmaceutical, and medical industries. Developing or optimizing culture media is essential to improve cell culture performance as a critical technology in cell culture engineering. Methodologies for media optimization have been developed to a great extent, such as the approaches of one-factor-at-a-time (OFAT) and response surface methodology (RSM). The present review introduces the emerging machine learning (ML) technology in cell culture engineering by combining high-throughput experimental technologies to develop highly efficient and effective culture media. The commonly used ML algorithms and the successful applications of employing ML in medium optimization are summarized. This review highlights the benefits of ML-assisted medium development and guides the selection of the media optimization method appropriate for various cell culture purposes.
Collapse
Affiliation(s)
- Takamasa Hashizume
- School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8572 Ibaraki, Japan
| | - Bei-Wen Ying
- School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8572 Ibaraki, Japan.
| |
Collapse
|
16
|
Niemis W, Peterson SR, Javier C, Nguyen A, Subiah S, Palmer RHC. On the utilization of the induced pluripotent stem cell (iPSC) model to study substance use disorders: A scoping review protocol. PLoS One 2023; 18:e0292238. [PMID: 37824561 PMCID: PMC10569547 DOI: 10.1371/journal.pone.0292238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 09/13/2023] [Indexed: 10/14/2023] Open
Abstract
INTRODUCTION Induced pluripotent stem cells (iPSCs) are cells derived from somatic cells via reprogramming techniques. The iPSC approach has been increasingly used in neuropsychiatric research in the last decade. Though substance use disorders (SUDs) are a commonly occurring psychiatric disorder, the application of iPSC model in addiction research has been limited. No comprehensive review has been reported. We conducted a scoping review to collate existing evidence on the iPSC technologies applied to SUD research. We aim to identify current knowledge gaps and limitations in order to advance the use of iPSCs in the SUD field. METHODS AND ANALYSIS We employed a scoping review using the methodological framework first created by Arksey and O'Malley and further updated by Levac et al. and the Joanna Briggs Institute (JBI). We adopted the Preferred Reporting Items for Systematic reviews and Meta-Analyses extension for Protocols (PRISMA-P) to report items for the protocol. We searched evidence from four electronic databases: PubMed®, Embase®, Web of Science™, and Scopus®. Primary research, systematic reviews, and meta-analyses were included and limited to studies published in English, at the time from 2007 to March 2022. This is an "ongoing" scoping review. Searched studies will be independently screened, selected, and extracted by two reviewers. Disagreement will be solved by the third reviewer and discussion. Extracted data will be analyzed in descriptive and quantitative approaches, then summarized and presented in appropriate formats. Results will be reported following the Preferred Reporting Items for Systematic reviews and Meta-Analyses extension for Scoping Reviews (PRISMA-ScR) guideline and disseminated through a peer-reviewed publication and conference presentations. CONCLUSION To our best knowledge, this is the first comprehensive scoping review of iPSC methods specifically applied to a broad range of addictive drugs/substances that lead to SUDs or misuse behavior. REGISTRATION This protocol is registered on Zenodo repository (https://zenodo.org/) with doi:10.5281/zenodo.7915252.
Collapse
Affiliation(s)
- Wasiri Niemis
- Behavioral Genetics of Addiction Laboratory, Department of Psychology, Emory University, Atlanta, GA, United States of America
| | - Shenita R. Peterson
- Woodruff Health Sciences Center Library, Emory University, Atlanta, GA, United States of America
| | - Chrisabella Javier
- Behavioral Genetics of Addiction Laboratory, Department of Psychology, Emory University, Atlanta, GA, United States of America
| | - Amy Nguyen
- Behavioral Genetics of Addiction Laboratory, Department of Psychology, Emory University, Atlanta, GA, United States of America
| | - Sanchi Subiah
- Behavioral Genetics of Addiction Laboratory, Department of Psychology, Emory University, Atlanta, GA, United States of America
| | - Rohan H. C. Palmer
- Behavioral Genetics of Addiction Laboratory, Department of Psychology, Emory University, Atlanta, GA, United States of America
| |
Collapse
|
17
|
Cheng YS, Xu M, Chen G, Beers J, Chen CZ, Liu C, Zou J, Zheng W. A Protocol for Culture and Characterization of Human Induced Pluripotent Stem Cells After Induction. Curr Protoc 2023; 3:e866. [PMID: 37610273 PMCID: PMC10506163 DOI: 10.1002/cpz1.866] [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] [Indexed: 08/24/2023]
Abstract
Human induced pluripotent stem cells (hiPSCs) are characterized by unlimited self-renewal and the capability to differentiate into all three germ layers, with the potential to further differentiate into all types of cells and tissues. Human iPSCs retain all genetic information from their original donors and can be developed into disease models to study disease pathophysiology, identify disease phenotypes and biomarkers, and evaluate therapeutic efficacy and toxicity for drug development. Human iPSCs can also be used to develop cell therapies and regenerative medicine. In the last decade, the technologies for hiPSC generation and differentiation have advanced rapidly. Human iPSC culture and propagation are tedious and require careful handling. High-quality hiPSCs are necessary for downstream applications. The methods, techniques, and skills for hiPSC maintenance and characterization are very different from those for immortalized cell lines. It can be a challenge for new laboratory staff, and sometimes even for experienced staff, to properly culture and maintain the high quality of these cells. Here, we describe a comprehensive set of protocols for hiPSC propagation under chemically defined and feeder-free culture conditions. These step-by-step protocols describe in detail all the reagents and experimental procedures needed to culture hiPSCs. The protocols also describe experimental methods for hiPSC characterization, including immunofluorescence staining and flow cytometric analysis with a panel of pluripotency markers, a teratoma formation assay for validation of in vivo pluripotency, and detection of Sendai virus to ensure elimination of the viral vectors. These protocols have been successfully used in our laboratory for hiPSC expansion and propagation, and this article provide a useful reference guide for laboratory staff to work on hiPSC culture. Published 2023. This article is a U.S. Government work and is in the public domain in the USA. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Propagation and cryopreservation of hiPSC cultures Basic Protocol 2: Recovery of cryopreserved hiPSCs Basic Protocol 3: Validation of pluripotency markers via immunocytochemical analysis Alternate Protocol: Determination of the expression of pluripotency markers via flow cytometry analysis Basic Protocol 4: Assessment of pluripotency via in vivo teratoma formation assay Basic Protocol 5: Confirmation of Sendai viral vector clearance via RT-PCR.
Collapse
Affiliation(s)
- Yu-Shan Cheng
- National Center for Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD 20892-3375, USA
| | - Miao Xu
- National Center for Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD 20892-3375, USA
| | - Guibin Chen
- National Center for Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD 20892-3375, USA
| | - Jeanette Beers
- iPSC Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892-3375, USA
| | - Catherine Z. Chen
- National Center for Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD 20892-3375, USA
| | - Chengyu Liu
- Transgenic Core, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892-3375, USA
| | - Jizhong Zou
- iPSC Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892-3375, USA
| | - Wei Zheng
- National Center for Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD 20892-3375, USA
| |
Collapse
|
18
|
Gao T, Zhao X, Hao J, Tian Y, Ma H, Liu W, An B, Sun F, Liu S, Guo B, Niu S, Li Z, Wang C, Wang Y, Feng G, Wang L, Li W, Wu J, Guo M, Zhou Q, Gu Q. A scalable culture system incorporating microcarrier for specialised mesenchymal stem cells from human embryonic stem cells. Mater Today Bio 2023; 20:100662. [PMID: 37214547 PMCID: PMC10196860 DOI: 10.1016/j.mtbio.2023.100662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/20/2023] [Accepted: 05/05/2023] [Indexed: 05/24/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) derived from human embryonic stem cells (hESCs) are a desirable cell source for cell therapy owing to their capacity to be produced stably and homogeneously in large quantities. However, a scalable culture system for hPSC-derived MSCs is urgently needed to meet the cell quantity and quality requirements of practical clinical applications. In this study, we developed a new microcarrier with hyaluronic acid (HA) as the core material, which allowed scalable serum-free suspension culture of hESC-derived MSCs (IMRCs). We used optimal microcarriers with a coating collagen concentration of 100 μg/mL or concave-structured surface (cHAMCs) for IMRC amplification in a stirred bioreactor, expanding IMRCs within six days with the highest yield of over one million cells per milliliter. In addition, the harvested cells exhibited high viability, immunomodulatory and regenerative therapeutic promise comparable to monolayer cultured MSCs while showing more increased secretion of extracellular matrix (ECM), particularly collagen-related proteins. In summary, we have established a scalable culture system for hESC-MSCs, providing novel approaches for future cell therapies.
Collapse
Affiliation(s)
- Tingting Gao
- State Key Laboratory of Stem Cell and Reproductive Biology, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiyuan Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jie Hao
- National Stem Cell Resource Center, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yao Tian
- State Key Laboratory of Stem Cell and Reproductive Biology, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Huike Ma
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Wenjing Liu
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Bin An
- State Key Laboratory of Stem Cell and Reproductive Biology, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Faguo Sun
- State Key Laboratory of Stem Cell and Reproductive Biology, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shasha Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Baojie Guo
- State Key Laboratory of Stem Cell and Reproductive Biology, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuaishuai Niu
- National Stem Cell Resource Center, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Zhongwen Li
- State Key Laboratory of Stem Cell and Reproductive Biology, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Chenxin Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Yukai Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guihai Feng
- State Key Laboratory of Stem Cell and Reproductive Biology, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Liu Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jun Wu
- State Key Laboratory of Stem Cell and Reproductive Biology, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Meijin Guo
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Qi Zhou
- State Key Laboratory of Stem Cell and Reproductive Biology, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qi Gu
- State Key Laboratory of Stem Cell and Reproductive Biology, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
19
|
Mazinani M, Rahbarizadeh F. New cell sources for CAR-based immunotherapy. Biomark Res 2023; 11:49. [PMID: 37147740 PMCID: PMC10163725 DOI: 10.1186/s40364-023-00482-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 04/04/2023] [Indexed: 05/07/2023] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy, in which a patient's own T lymphocytes are engineered to recognize and kill cancer cells, has achieved striking success in some hematological malignancies in preclinical and clinical trials, resulting in six FDA-approved CAR-T products currently available in the market. Despite impressive clinical outcomes, concerns about treatment failure associated with low efficacy or high cytotoxicity of CAR-T cells remain. While the main focus has been on improving CAR-T cells, exploring alternative cellular sources for CAR generation has garnered growing interest. In the current review, we comprehensively evaluated other cell sources rather than conventional T cells for CAR generation.
Collapse
Affiliation(s)
- Marzieh Mazinani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, P.O. Box 14115-111, Tehran, Iran
| | - Fatemeh Rahbarizadeh
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, P.O. Box 14115-111, Tehran, Iran.
- Research and Development Center of Biotechnology, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
20
|
Zhang Y, Lyu H, Guo R, Cao X, Feng J, Jin X, Lu W, Zhao M. Epstein‒Barr virus-associated cellular immunotherapy. Cytotherapy 2023:S1465-3249(23)00099-3. [PMID: 37149797 DOI: 10.1016/j.jcyt.2023.04.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 03/24/2023] [Accepted: 04/10/2023] [Indexed: 05/08/2023]
Abstract
Epstein‒Barr virus (EBV) is a human herpes virus that is saliva-transmissible and universally asymptomatic. It has been confirmed that more than 90% of the population is latently infected with EBV for life. EBV can cause a variety of related cancers, such as nasopharyngeal carcinoma, diffuse large B-cell lymphoma, and Burkitt lymphoma. Currently, many clinical studies have demonstrated that EBV-specific cytotoxic T lymphocytes and other cell therapies can be safely and effectively transfused to prevent and treat some diseases caused by EBV. This review will mainly focus on discussing EBV-specific cytotoxic T lymphocytes and will touch on therapeutic EBV vaccines and chimeric antigen receptor T-cell therapy briefly.
Collapse
Affiliation(s)
- Yi Zhang
- First Center Clinic College of Tianjin Medical University, Tianjin, China.
| | - Hairong Lyu
- Department of Hematology, Tianjin First Central Hospital, Tianjin, China
| | - Ruiting Guo
- First Center Clinic College of Tianjin Medical University, Tianjin, China
| | - Xinping Cao
- First Center Clinic College of Tianjin Medical University, Tianjin, China
| | - Juan Feng
- Tianjin Jizhou District People's Hospital, Tianjin, China
| | - Xin Jin
- Department of Hematology, Tianjin First Central Hospital, Tianjin, China
| | - Wenyi Lu
- Department of Hematology, Tianjin First Central Hospital, Tianjin, China.
| | - Mingfeng Zhao
- Department of Hematology, Tianjin First Central Hospital, Tianjin, China.
| |
Collapse
|
21
|
Aguirre-Vázquez A, Castorena-Torres F, Silva-Ramírez B, Peñuelas-Urquides K, Camacho-Moll ME, Salazar-Olivo LA, Velasco I, Bermúdez de León M. Cell-type dependent regulation of pluripotency and chromatin remodeling genes by hydralazine. Stem Cell Res Ther 2023; 14:42. [PMID: 36927767 PMCID: PMC10021945 DOI: 10.1186/s13287-023-03268-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 03/08/2023] [Indexed: 03/18/2023] Open
Abstract
BACKGROUND The generation of induced pluripotent stem cells has opened the field of study for stem cell research, disease modeling and drug development. However, the epigenetic signatures present in somatic cells make cell reprogramming still an inefficient process. This epigenetic memory constitutes an obstacle in cellular reprogramming. Here, we report the effect of hydralazine (HYD) and valproic acid (VPA), two small molecules with proven epigenetic activity, on the expression of pluripotency genes in adult (aHF) and neonatal (nbHF) human fibroblasts. METHODS aHF and nbHF were treated with HYD and/or VPA, and viability and gene expression assays for OCT4, NANOG, c-MYC, KLF4, DNMT1, TET3, ARID1A and ARID2 by quantitative PCR were performed. aHF and nbHF were transfected with episomal plasmid bearing Yamanaka factors (OCT4, SOX2, KLF4 and c-MYC) and exposed to HYD and VPA to determine the reprogramming efficiency. Methylation sensitive restriction enzyme (MSRE) qPCR assays were performed on OCT4 and NANOG promoter regions. Immunofluorescence assays were carried out for pluripotency genes on iPSC derived from aHF and nbHF. RESULTS HYD upregulated the expression of OCT4 (2.5-fold) and NANOG (fourfold) genes but not c-Myc or KLF4 in aHF and had no significant effect on the expression of all these genes in nbHF. VPA upregulated the expression of NANOG (twofold) in aHF and c-MYC in nbHF, while it downregulated the expression of NANOG in nbHF. The combination of HYD and VPA canceled the OCT4 and NANOG overexpression induced by HYD in aHF, while it reinforced the effects of VPA on c-Myc expression in nbHF. The HYD-induced overexpression of OCT4 and NANOG in aHDF was not dependent on demethylation of gene promoters, and no changes in the reprogramming efficiency were observed in both cell populations despite the downregulation of epigenetic genes DNMT1, ARID1A, and ARID2 in nbHF. CONCLUSIONS Our data provide evidence that HYD regulates the expression of OCT4 and NANOG pluripotency genes as well as ARID1A and ARID2 genes, two members of the SWI/SNF chromatin remodeling complex family, in normal human dermal fibroblasts.
Collapse
Affiliation(s)
- Alain Aguirre-Vázquez
- Departamento de Biología Molecular, Centro de Investigación Biomédica del Noreste, Instituto Mexicano del Seguro Social, 64720, Monterrey, Nuevo León, Mexico.,Depto. de Biología Molecular, Instituto Potosino de Investigación Científica y Tecnológica, 78216, San Luis Potosí, S.L.P., Mexico
| | | | - Beatriz Silva-Ramírez
- Departamento de Inmunogenética, Centro de Investigación Biomédica del Noreste, Instituto Mexicano del Seguro Social, 64720, Monterrey, Nuevo León, Mexico
| | - Katia Peñuelas-Urquides
- Departamento de Biología Molecular, Centro de Investigación Biomédica del Noreste, Instituto Mexicano del Seguro Social, 64720, Monterrey, Nuevo León, Mexico
| | - María Elena Camacho-Moll
- Departamento de Biología Molecular, Centro de Investigación Biomédica del Noreste, Instituto Mexicano del Seguro Social, 64720, Monterrey, Nuevo León, Mexico
| | - Luis A Salazar-Olivo
- Depto. de Biología Molecular, Instituto Potosino de Investigación Científica y Tecnológica, 78216, San Luis Potosí, S.L.P., Mexico
| | - Iván Velasco
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico.,Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", 14269, Mexico City, Mexico
| | - Mario Bermúdez de León
- Departamento de Biología Molecular, Centro de Investigación Biomédica del Noreste, Instituto Mexicano del Seguro Social, 64720, Monterrey, Nuevo León, Mexico.
| |
Collapse
|
22
|
Hattab D, Amer MFA, Mohd Gazzali A, Chuah LH, Bakhtiar A. Current status in cellular-based therapies for prevention and treatment of COVID-19. Crit Rev Clin Lab Sci 2023:1-25. [PMID: 36825325 DOI: 10.1080/10408363.2023.2177605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the pathogen responsible for the coronavirus disease 2019 (COVID-19) outbreaks that resulted in a catastrophic threat to global health, with more than 500 million cases detected and 5.5 million deaths worldwide. Patients with a COVID-19 infection presented with clinical manifestations ranging from asymptomatic to severe symptoms, resulting in acute lung injury, acute respiratory distress syndrome, and even death. Immune dysregulation through delayed innate immune response or impairment of the adaptive immune response is the key contributor to the pathophysiology of COVID-19 and SARS-CoV-2-induced cytokine storm. Symptomatic and supportive therapy is the fundamental strategy in treating COVID-19 infection, including antivirals, steroid-based therapies, and cell-based immunotherapies. Various studies reported substantial effects of immune-based therapies for patients with COVID-19 to modulate the over-activated immune system while simultaneously refining the body's ability to destroy the virus. However, challenges may arise from the complexity of the disease through the genetic variance of the virus itself and patient heterogeneity, causing increased transmissibility and heightened immune system evasion that rapidly change the intervention and prevention measures for SARS-CoV-2. Cell-based therapy, utilizing stem cells, dendritic cells, natural killer cells, and T cells, among others, are being extensively explored as other potential immunological approaches for preventing and treating SARS-CoV-2-affected patients the similar process was effectively proven in SARS-CoV-1 and MERS-CoV infections. This review provides detailed insights into the innate and adaptive immune response-mediated cell-based immunotherapies in COVID-19 patients. The immune response linking towards engineered autologous or allogenic immune cells for either treatment or preventive therapies is subsequently highlighted in an individual study or in combination with several existing treatments. Up-to-date data on completed and ongoing clinical trials of cell-based agents for preventing or treating COVID-19 are also outlined to provide a guide that can help in treatment decisions and future trials.
Collapse
Affiliation(s)
- Dima Hattab
- Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Mumen F A Amer
- Faculty of Pharmacy, Applied Science Private University, Amman, Jordan
| | - Amirah Mohd Gazzali
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Penang, Malaysia
| | - Lay Hong Chuah
- School of Pharmacy, Monash University Malaysia, Selangor, Malaysia
| | - Athirah Bakhtiar
- School of Pharmacy, Monash University Malaysia, Selangor, Malaysia
| |
Collapse
|
23
|
Han H, Rim YA, Ju JH. Recent updates of stem cell-based erythropoiesis. Hum Cell 2023; 36:894-907. [PMID: 36754940 PMCID: PMC9908308 DOI: 10.1007/s13577-023-00872-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 01/28/2023] [Indexed: 02/10/2023]
Abstract
Blood transfusions are now an essential part of modern medicine. Transfusable red blood cells (RBCs) are employed in various therapeutic strategies; however, the processes of blood donation, collection, and administration still involve many limitations. Notably, a lack of donors, the risk of transfusion-transmitted disease, and recent pandemics such as COVID-19 have prompted us to search for alternative therapeutics to replace this resource. Originally, RBC production was attempted via the ex vivo differentiation of stem cells. However, a more approachable and effective cell source is now required for broader applications. As a viable alternative, pluripotent stem cells have been actively used in recent research. In this review, we discuss the basic concepts related to erythropoiesis, as well as early research using hematopoietic stem cells ex vivo, and discuss the current trend of in vitro erythropoiesis using human-induced pluripotent stem cells.
Collapse
Affiliation(s)
- Heeju Han
- Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, , Seoul, Republic of Korea ,Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yeri Alice Rim
- Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| | - Ji Hyeon Ju
- Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea. .,Division of Rheumatology, Department of Internal Medicine, Institute of Medical Science, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
24
|
Kim M, Park J, Kim S, Han DW, Shin B, Schöler HR, Kim J, Kim KP. Generation of Induced Pluripotent Stem Cells from Lymphoblastoid Cell Lines by Electroporation of Episomal Vectors. Int J Stem Cells 2022; 16:36-43. [PMID: 36581370 PMCID: PMC9978833 DOI: 10.15283/ijsc22177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 10/31/2022] [Accepted: 11/03/2022] [Indexed: 12/31/2022] Open
Abstract
Background and Objectives Lymphoblastoid cell lines (LCLs) deposited from disease-affected individuals could be a valuable donor cell source for generating disease-specific induced pluripotent stem cells (iPSCs). However, generation of iPSCs from the LCLs is still challenging, as yet no effective gene delivery strategy has been developed. Methods and Results Here, we reveal an effective gene delivery method specifically for LCLs. We found that LCLs appear to be refractory toward retroviral and lentiviral transduction. Consequently, lentiviral and retroviral transduction of OCT4, SOX2, KFL4 and c-MYC into LCLs does not elicit iPSC colony formation. Interestingly, however we found that transfection of oriP/EBNA-1-based episomal vectors by electroporation is an efficient gene delivery system into LCLs, enabling iPSC generation from LCLs. These iPSCs expressed pluripotency makers (OCT4, NANOG, SSEA4, SALL4) and could form embryoid bodies. Conclusions Our data show that electroporation is an effective gene delivery method with which LCLs can be efficiently reprogrammed into iPSCs.
Collapse
Affiliation(s)
- Myunghyun Kim
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Junmyeong Park
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sujin Kim
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Dong Wook Han
- School of Biotechnology and Healthcare, Wuyi University, Jiangmen, China
| | - Borami Shin
- Department of General Pediatrics, University of Children’s Hospital Muenster, Muenster, Germany
| | - Hans Robert Schöler
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Johnny Kim
- Department of Cardiac Development and Remodelling, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Kee-Pyo Kim
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, Korea,Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany,Correspondence to Kee-Pyo Kim, Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Korea, Tel: +82-2-3147-8409, Fax: +82-2-532-9575, E-mail:
| |
Collapse
|
25
|
Long-term calcium imaging reveals functional development in hiPSC-derived cultures comparable to human but not rat primary cultures. Stem Cell Reports 2022; 18:205-219. [PMID: 36563684 PMCID: PMC9860124 DOI: 10.1016/j.stemcr.2022.11.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 11/15/2022] [Accepted: 11/18/2022] [Indexed: 12/24/2022] Open
Abstract
Models for human brain-oriented research are often established on primary cultures from rodents, which fails to recapitulate cellular specificity and molecular cues of the human brain. Here we investigated whether neuronal cultures derived from human induced pluripotent stem cells (hiPSCs) feature key advantages compared with rodent primary cultures. Using calcium fluorescence imaging, we tracked spontaneous neuronal activity in hiPSC-derived, human, and rat primary cultures and compared their dynamic and functional behavior as they matured. We observed that hiPSC-derived cultures progressively changed upon development, exhibiting gradually richer activity patterns and functional traits. By contrast, rat primary cultures were locked in the same dynamic state since activity onset. Human primary cultures exhibited features in between hiPSC-derived and rat primary cultures, although traits from the former predominated. Our study demonstrates that hiPSC-derived cultures are excellent models to investigate development in neuronal assemblies, a hallmark for applications that monitor alterations caused by damage or neurodegeneration.
Collapse
|
26
|
Hann SY, Cui H, Chen G, Boehm M, Esworthy T, Zhang LG. 3D printed biomimetic flexible blood vessels with iPS cell-laden hierarchical multilayers. BIOMEDICAL ENGINEERING ADVANCES 2022; 4:100065. [PMID: 36582411 PMCID: PMC9794201 DOI: 10.1016/j.bea.2022.100065] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Successful recovery from vascular diseases has typically relied on the surgical repair of damaged blood vessels (BVs), with the majority of current approaches involving the implantation of autologous BVs, which is plagued by donor site tissue damage. Researchers have attempted to develop artificial vessels as an alternative solution to traditional approaches to BV repair. However, the manufacturing of small-diameter (< 6 mm) BVs is still considered one of the biggest challenges due to its difficulty in the precise fabrication and the replication of biomimetic architectures. In this study, we successfully developed 3D printed flexible small-diameter BVs that consist of smooth muscle cells and a vascularized endothelium. In the developed artificial BV, a rubber-like elastomer was printed as the outermost layer of the vessel, which demonstrated enhanced mechanical properties, while and human induced pluripotent stem cell (iPSC)-derived vascular smooth muscle cells (iSMCs) and endothelial cells (iECs) embedded fibrinogen solutions were coaxially extruded with thrombin solution to form cell-laden fibrin gel inner layers. Our results showed that the 3D BVs possessed proper mechanical properties, and the cells in the fibrin layers substantially proliferated over time to form a stable BV construct. Our study demonstrated that the 3D printed flexible small-diameter BV using iPSCs could be a promising platform for the treatment of vascular diseases.
Collapse
Affiliation(s)
- Sung Yun Hann
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
| | - Haitao Cui
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
| | - Guibin Chen
- Laboratory of Cardiovascular Regenerative Medicine, Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Manfred Boehm
- Laboratory of Cardiovascular Regenerative Medicine, Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Timothy Esworthy
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
| | - Lijie Grace Zhang
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
- Department of Electrical and Computer Engineering, The George Washington University, Washington, DC 20052, USA
- Department of Biomedical Engineering, The George Washington University, Washington, DC 20052, USA
- Department of Medicine, The George Washington University Medical Center, Washington, DC 20052, USA
| |
Collapse
|
27
|
Abdelsayed M, Kort EJ, Jovinge S, Mercola M. Repurposing drugs to treat cardiovascular disease in the era of precision medicine. Nat Rev Cardiol 2022; 19:751-764. [PMID: 35606425 PMCID: PMC9125554 DOI: 10.1038/s41569-022-00717-6] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/22/2022] [Indexed: 12/14/2022]
Abstract
Drug repurposing is the use of a given therapeutic agent for indications other than that for which it was originally designed or intended. The concept is appealing because of potentially lower development costs and shorter timelines than are needed to produce a new drug. To date, drug repurposing for cardiovascular indications has been opportunistic and driven by knowledge of disease mechanisms or serendipitous observation rather than by systematic endeavours to match an existing drug to a new indication. Innovations in two areas of personalized medicine - computational approaches to associate drug effects with disease signatures and predictive model systems to screen drugs for disease-modifying activities - support efforts that together create an efficient pipeline to systematically repurpose drugs to treat cardiovascular disease. Furthermore, new experimental strategies that guide the medicinal chemistry re-engineering of drugs could improve repurposing efforts by tailoring a medicine to its new indication. In this Review, we summarize the historical approach to repurposing and discuss the technological advances that have created a new landscape of opportunities.
Collapse
Affiliation(s)
- Mena Abdelsayed
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Department of Medicine, Stanford University, Stanford, CA, USA
| | - Eric J Kort
- DeVos Cardiovascular Program Spectrum Health & Van Andel Institute, Grand Rapids, MI, USA
| | - Stefan Jovinge
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA.
- DeVos Cardiovascular Program Spectrum Health & Van Andel Institute, Grand Rapids, MI, USA.
- Department of Medicine, University of Texas Southwestern, Dallas, TX, USA.
- Department of Clinical Sciences, Scania University Hospital, Lund University, Lund, Sweden.
| | - Mark Mercola
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA.
- Department of Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
28
|
Cao X, Weil MM, Wu JC. Clinical Trial in a Dish for Space Radiation Countermeasure Discovery. LIFE SCIENCES IN SPACE RESEARCH 2022; 35:140-149. [PMID: 36336359 PMCID: PMC10947779 DOI: 10.1016/j.lssr.2022.05.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/30/2022] [Accepted: 05/25/2022] [Indexed: 06/16/2023]
Abstract
NASA aims to return humans to the moon within the next five years and to land humans on Mars in a few decades. Space radiation exposure represents a major challenge to astronauts' health during long-duration missions, as it is linked to increased risks of cancer, cardiovascular dysfunctions, central nervous system (CNS) impairment, and other negative outcomes. Characterization of radiation health effects and developing corresponding countermeasures are high priorities for the preparation of long duration space travel. Due to limitations of animal and cell models, the development of novel physiologically relevant radiation models is needed to better predict these individual risks and bridge gaps between preclinical testing and clinical trials in drug development. "Clinical Trial in a Dish" (CTiD) is now possible with the use of human induced pluripotent stem cells (hiPSCs), offering a powerful tool for drug safety or efficacy testing using patient-specific cell models. Here we review the development and applications of CTiD for space radiation biology and countermeasure studies, focusing on progress made in the past decade.
Collapse
Affiliation(s)
- Xu Cao
- Stanford Cardiovascular Institute, Stanford University School of Medicine, CA 94305, USA; Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael M Weil
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, CA 94305, USA; Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Radiology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
29
|
Labau JIR, Andelic M, Faber CG, Waxman SG, Lauria G, Dib-Hajj SD. Recent advances for using human induced-pluripotent stem cells as pain-in-a-dish models of neuropathic pain. Exp Neurol 2022; 358:114223. [PMID: 36100046 DOI: 10.1016/j.expneurol.2022.114223] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 08/15/2022] [Accepted: 09/05/2022] [Indexed: 11/28/2022]
Abstract
Neuropathic pain is amongst the most common non-communicable disorders and the poor effectiveness of current treatment is an unmet need. Although pain is a universal experience, there are significant inter-individual phenotypic differences. Developing models that can accurately recapitulate the clinical pain features is crucial to better understand underlying pathophysiological mechanisms and find innovative treatments. Current data from heterologous expression systems that investigate properties of specific molecules involved in pain signaling, and from animal models, show limited success with their translation into the development of novel treatments for pain. This is in part because they do not recapitulate the native environment in which a particular molecule functions, and due to species-specific differences in the properties of several key molecules that are involved in pain signaling. The limited availability of post-mortem tissue, in particular dorsal root ganglia (DRG), has hampered research using human cells in pre-clinical studies. Human induced-pluripotent stem cells (iPSCs) have emerged as an exciting alternative platform to study patient-specific diseases. Sensory neurons that are derived from iPSCs (iPSC-SNs) have provided new avenues towards elucidating peripheral pathophysiological mechanisms, the potential for development of personalized treatments, and as a cell-based system for high-throughput screening for discovering novel analgesics. Nevertheless, reprogramming and differentiation protocols to obtain nociceptors have mostly yielded immature homogenous cell populations that do not recapitulate the heterogeneity of native sensory neurons. To close the gap between native human tissue and iPSCs, alternative strategies have been developed. We will review here recent developments in differentiating iPSC-SNs and their use in pre-clinical translational studies. Direct conversion of stem cells into the cells of interest has provided a more cost- and time-saving method to improve reproducibility and diversity of sensory cell types. Furthermore, multi-cellular strategies that mimic in vivo microenvironments for cell maturation, by improving cell contact and communication (co-cultures), reproducing the organ complexity and architecture (three-dimensional organoid), and providing iPSCs with the full spatiotemporal context and nutrients needed for acquiring a mature phenotype (xenotransplantation), have led to functional sensory neuron-like systems. Finally, this review touches on novel prospective strategies, including fluorescent-tracking to select the differentiated neurons of relevance, and dynamic clamp, an electrophysiological method that allows direct manipulation of ionic conductances that are missing in iPSC-SNs.
Collapse
Affiliation(s)
- Julie I R Labau
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA; Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA; Department of Toxicogenomics, Clinical Genomics, Maastricht University Medical Centre+, Maastricht, the Netherlands; School of Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Mirna Andelic
- School of Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands; Department of Neurology, Neuroalgology Unit, Fondazione IRCCS Istituto Neurologico "Carlo Besta", Milan, Italy
| | - Catharina G Faber
- School of Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA; Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA
| | - Giuseppe Lauria
- Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico "Carlo Besta", Milan, Italy; Department of Medical Biotechnology and Translational Medicine, University of Milan, Italy.
| | - Sulayman D Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA; Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA.
| |
Collapse
|
30
|
Xu T, Duan J, Li Y, Wang G, Li S, Li Y, Lu W, Yan X, Ren Y, Guo F, Cao L, Lu J. Generation of a TPH2-EGFP reporter cell line for purification and monitoring of human serotonin neurons in vitro and in vivo. Stem Cell Reports 2022; 17:2365-2379. [PMID: 36150384 PMCID: PMC9561537 DOI: 10.1016/j.stemcr.2022.08.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 08/24/2022] [Accepted: 08/25/2022] [Indexed: 10/25/2022] Open
Abstract
Generation of serotonin neurons (SNs) from human pluripotent stem cells (hPSCs) provides a promising platform to explore the mechanisms of serotonin-associated neuropsychiatric disorders. However, neural differentiation always yields heterogeneous cell populations, making it difficult to identify and purify SNs in vitro or track them in vivo following transplantation. Herein, we generated a TPH2-EGFP reporter hPSC line with insertion of EGFP into the endogenous tryptophan hydroxylase 2 (TPH2) locus using CRISPR-Cas9-mediated gene editing technology. This TPH2-reporter, which faithfully indicated TPH2 expression during differentiation, enabled us to obtain purified SNs for subsequent transcriptional analysis and study of pharmacological responses to antidepressants. In addition, the reporter system showed strong EGFP expression to indicate SNs, which enabled us to explore in vitro and ex vivo electrophysiological properties of SNs. In conclusion, this TPH2-EGFP reporter cell line might be of great significance for studies on human SN-related development and differentiation, drug screening, disease modeling, and cell replacement therapies.
Collapse
Affiliation(s)
- 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 200092, China
| | - Jinjin Duan
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; School of Life Sciences, Shanghai University, Shanghai 200444, 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 200092, China
| | - 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 200092, 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 200092, China
| | - You 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 200092, China
| | - Wenting 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 200092, China
| | - Xinyi Yan
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Yixuan Ren
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Fei Guo
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, 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 200092, China.
| | - 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 200092, China; Suzhou Institute of Tongji University, Suzhou 215101, China.
| |
Collapse
|
31
|
Tarricone G, Carmagnola I, Chiono V. Tissue-Engineered Models of the Human Brain: State-of-the-Art Analysis and Challenges. J Funct Biomater 2022; 13:146. [PMID: 36135581 PMCID: PMC9501967 DOI: 10.3390/jfb13030146] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/03/2022] [Accepted: 09/06/2022] [Indexed: 11/26/2022] Open
Abstract
Neurological disorders affect billions of people across the world, making the discovery of effective treatments an important challenge. The evaluation of drug efficacy is further complicated because of the lack of in vitro models able to reproduce the complexity of the human brain structure and functions. Some limitations of 2D preclinical models of the human brain have been overcome by the use of 3D cultures such as cell spheroids, organoids and organs-on-chip. However, one of the most promising approaches for mimicking not only cell structure, but also brain architecture, is currently represented by tissue-engineered brain models. Both conventional (particularly electrospinning and salt leaching) and unconventional (particularly bioprinting) techniques have been exploited, making use of natural polymers or combinations between natural and synthetic polymers. Moreover, the use of induced pluripotent stem cells (iPSCs) has allowed the co-culture of different human brain cells (neurons, astrocytes, oligodendrocytes, microglia), helping towards approaching the central nervous system complexity. In this review article, we explain the importance of in vitro brain modeling, and present the main in vitro brain models developed to date, with a special focus on the most recent advancements in tissue-engineered brain models making use of iPSCs. Finally, we critically discuss achievements, main challenges and future perspectives.
Collapse
Affiliation(s)
- Giulia Tarricone
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129 Turin, Italy
- PolitoBioMedLab, Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129 Turin, Italy
- Interuniversity Center for the Promotion of the 3Rs Principle in Teaching and Research, Centro 3R, 56122 Pisa, Italy
- Nanobiointeractions & Nanodiagnostics, Istituto Italiano di Tecnologia (IIT), Via Morego 30, 16163 Genova, Italy
- Department of Chemistry and Industrial Chemistry, University of Genova, Via Dodecaneso 31, 16146 Genova, Italy
| | - Irene Carmagnola
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129 Turin, Italy
- PolitoBioMedLab, Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129 Turin, Italy
- Interuniversity Center for the Promotion of the 3Rs Principle in Teaching and Research, Centro 3R, 56122 Pisa, Italy
| | - Valeria Chiono
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129 Turin, Italy
- PolitoBioMedLab, Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129 Turin, Italy
- Interuniversity Center for the Promotion of the 3Rs Principle in Teaching and Research, Centro 3R, 56122 Pisa, Italy
| |
Collapse
|
32
|
Nakashima Y, Yoshida S, Tsukahara M. Semi-3D cultures using Laminin 221 as a coating material for human induced pluripotent stem cells. Regen Biomater 2022; 9:rbac060. [PMID: 36176714 PMCID: PMC9514851 DOI: 10.1093/rb/rbac060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 07/09/2022] [Accepted: 08/21/2022] [Indexed: 11/19/2022] Open
Abstract
It was previously believed that human induced pluripotent stem cells (hiPSCs) did not show adhesion to the coating material Laminin 221, which is known to have specific affinity for cardiomyocytes. In this study, we report that human mononuclear cell-derived hiPSCs, established with Sendai virus vector, form peninsular-like colonies rather than embryonic stem cell-like colonies; these peninsular-like colonies can be passaged more than 10 times after establishment. Additionally, initialization-deficient cells with residual Sendai virus vector adhered to the coating material Laminin 511 but not to Laminin 221. Therefore, the expression of undifferentiated markers tended to be higher in hiPSCs established on Laminin 221 than on Laminin 511. On Laminin 221, hiPSCs15M66 showed a semi-floating colony morphology. The expression of various markers of cell polarity was significantly lower in hiPSCs cultured on Laminin 221 than in hiPSCs cultured on Laminin 511. Furthermore, 201B7 and 15M66 hiPSCs showed 3D cardiomyocyte differentiation on Laminin 221. Thus, the coating material Laminin 221 provides semi-floating culture conditions for the establishment, culture and induced differentiation of hiPSCs.
Collapse
Affiliation(s)
- Yoshiki Nakashima
- Kyoto University Center for iPS Cell Research and Application Foundation (CiRA Foundation), Facility for iPS Cell Therapy (FiT), Kyoto 606-8397, Japan
| | - Shinsuke Yoshida
- Kyoto University Center for iPS Cell Research and Application Foundation (CiRA Foundation), Facility for iPS Cell Therapy (FiT), Kyoto 606-8397, Japan
| | - Masayoshi Tsukahara
- Kyoto University Center for iPS Cell Research and Application Foundation (CiRA Foundation), Facility for iPS Cell Therapy (FiT), Kyoto 606-8397, Japan
| |
Collapse
|
33
|
Graffmann N, Adjaye J. Editorial for Special Issue: iPS Cells (iPSCs) for Modelling and Treatment of Human Diseases. Cells 2022; 11:2270. [PMID: 35892567 PMCID: PMC9332752 DOI: 10.3390/cells11152270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 07/21/2022] [Indexed: 02/01/2023] Open
Abstract
Human induced pluripotent stem cells (iPSCs) have evolved as a powerful tool to model diseases and study treatment possibilities [...].
Collapse
Affiliation(s)
- Nina Graffmann
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich-Heine University, 40225 Dusseldorf, Germany
| | - James Adjaye
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich-Heine University, 40225 Dusseldorf, Germany
| |
Collapse
|
34
|
Sim EZ, Enomoto T, Shiraki N, Furuta N, Kashio S, Kambe T, Tsuyama T, Arakawa A, Ozawa H, Yokoyama M, Miura M, Kume S. Methionine metabolism regulates pluripotent stem cell pluripotency and differentiation through zinc mobilization. Cell Rep 2022; 40:111120. [PMID: 35858556 DOI: 10.1016/j.celrep.2022.111120] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 04/19/2022] [Accepted: 06/28/2022] [Indexed: 11/03/2022] Open
Abstract
Pluripotent stem cells (PSCs) exhibit a unique feature that requires S-adenosylmethionine (SAM) for the maintenance of their pluripotency. Methionine deprivation in the medium causes a reduction in intracellular SAM, thus rendering PSCs in a state potentiated for differentiation. In this study, we find that methionine deprivation triggers a reduction in intracellular protein-bound Zn content and upregulation of Zn exporter SLC30A1 in PSCs. Culturing PSCs in Zn-deprived medium results in decreased intracellular protein-bound Zn content, reduced cell growth, and potentiated differentiation, which partially mimics methionine deprivation. PSCs cultured under Zn deprivation exhibit an altered methionine metabolism-related metabolite profile. We conclude that methionine deprivation potentiates differentiation partly by lowering cellular Zn content. We establish a protocol to generate functional pancreatic β cells by applying methionine and Zn deprivation. Our results reveal a link between Zn signaling and methionine metabolism in the regulation of cell fate in PSCs.
Collapse
Affiliation(s)
- Erinn Zixuan Sim
- School of Life Science and Technology, Tokyo Institute of Technology, 4259-B-25 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
| | - Takayuki Enomoto
- School of Life Science and Technology, Tokyo Institute of Technology, 4259-B-25 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
| | - Nobuaki Shiraki
- School of Life Science and Technology, Tokyo Institute of Technology, 4259-B-25 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan.
| | - Nao Furuta
- School of Life Science and Technology, Tokyo Institute of Technology, 4259-B-25 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
| | - Soshiro Kashio
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Taiho Kambe
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan
| | - Tomonori Tsuyama
- Division of Stem Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Akihiro Arakawa
- Research Institute for Bioscience Products and Fine Chemicals, Ajinomoto, Kawasaki-shi, Kanagawa, Japan
| | - Hiroki Ozawa
- School of Life Science and Technology, Tokyo Institute of Technology, 4259-B-25 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
| | - Mizuho Yokoyama
- Research Institute for Bioscience Products and Fine Chemicals, Ajinomoto, Kawasaki-shi, Kanagawa, Japan
| | - Masayuki Miura
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Shoen Kume
- School of Life Science and Technology, Tokyo Institute of Technology, 4259-B-25 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan.
| |
Collapse
|
35
|
Wang Z, McWilliams-Koeppen HP, Reza H, Ostberg JR, Chen W, Wang X, Huynh C, Vyas V, Chang WC, Starr R, Wagner JR, Aguilar B, Yang X, Wu X, Wang J, Chen W, Koelker-Wolfe E, Seet CS, Montel-Hagen A, Crooks GM, Forman SJ, Brown CE. 3D-organoid culture supports differentiation of human CAR+ iPSCs into highly functional CAR T cells. Cell Stem Cell 2022; 29:515-527.e8. [PMID: 35278370 PMCID: PMC9119152 DOI: 10.1016/j.stem.2022.02.009] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 09/10/2021] [Accepted: 02/14/2022] [Indexed: 12/13/2022]
Abstract
Unlimited generation of chimeric antigen receptor (CAR) T cells from human-induced pluripotent stem cells (iPSCs) is an attractive approach for "off-the-shelf" CAR T cell immunotherapy. Approaches to efficiently differentiate iPSCs into canonical αβ T cell lineages, while maintaining CAR expression and functionality, however, have been challenging. We report that iPSCs reprogramed from CD62L+ naive and memory T cells followed by CD19-CAR engineering and 3D-organoid system differentiation confers products with conventional CD8αβ-positive CAR T cell characteristics. Expanded iPSC CD19-CAR T cells showed comparable antigen-specific activation, degranulation, cytotoxicity, and cytokine secretion compared with conventional CD19-CAR T cells and maintained homogeneous expression of the TCR derived from the initial clone. iPSC CD19-CAR T cells also mediated potent antitumor activity in vivo, prolonging survival of mice with CD19+ human tumor xenografts. Our study establishes feasible methodologies to generate highly functional CAR T cells from iPSCs to support the development of "off-the-shelf" manufacturing strategies.
Collapse
Affiliation(s)
- Zhiqiang Wang
- Department of Hematology & Hematopoietic Cell Transplantation, T Cell Therapeutics Research Laboratories, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA.
| | - Helen P McWilliams-Koeppen
- Department of Hematology & Hematopoietic Cell Transplantation, T Cell Therapeutics Research Laboratories, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Hernan Reza
- Department of Hematology & Hematopoietic Cell Transplantation, T Cell Therapeutics Research Laboratories, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Julie R Ostberg
- Department of Hematology & Hematopoietic Cell Transplantation, T Cell Therapeutics Research Laboratories, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Wuyang Chen
- Department of Hematology & Hematopoietic Cell Transplantation, T Cell Therapeutics Research Laboratories, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Xiuli Wang
- Department of Hematology & Hematopoietic Cell Transplantation, T Cell Therapeutics Research Laboratories, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Christian Huynh
- Department of Hematology & Hematopoietic Cell Transplantation, T Cell Therapeutics Research Laboratories, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Vibhuti Vyas
- Department of Hematology & Hematopoietic Cell Transplantation, T Cell Therapeutics Research Laboratories, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Wen-Chung Chang
- Department of Hematology & Hematopoietic Cell Transplantation, T Cell Therapeutics Research Laboratories, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Renate Starr
- Department of Hematology & Hematopoietic Cell Transplantation, T Cell Therapeutics Research Laboratories, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Jamie R Wagner
- Department of Hematology & Hematopoietic Cell Transplantation, T Cell Therapeutics Research Laboratories, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Brenda Aguilar
- Department of Hematology & Hematopoietic Cell Transplantation, T Cell Therapeutics Research Laboratories, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Xin Yang
- Department of Hematology & Hematopoietic Cell Transplantation, T Cell Therapeutics Research Laboratories, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Xiwei Wu
- Integrative Genomics Core, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Jinhui Wang
- Integrative Genomics Core, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Wei Chen
- Integrative Genomics Core, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Ellery Koelker-Wolfe
- Department of Hematology & Hematopoietic Cell Transplantation, T Cell Therapeutics Research Laboratories, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Christopher S Seet
- Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA; Broad Stem Cell Research Center, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Amélie Montel-Hagen
- Department of Pathology & Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Gay M Crooks
- Broad Stem Cell Research Center, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA; Department of Pathology & Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA; Division of Pediatric Hematology-Oncology, Department of Pediatrics, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Stephen J Forman
- Department of Hematology & Hematopoietic Cell Transplantation, T Cell Therapeutics Research Laboratories, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Christine E Brown
- Department of Hematology & Hematopoietic Cell Transplantation, T Cell Therapeutics Research Laboratories, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA.
| |
Collapse
|
36
|
The role of metabolism in directed differentiation versus trans-differentiation of cardiomyocytes. Semin Cell Dev Biol 2022; 122:56-65. [PMID: 34074592 PMCID: PMC8725317 DOI: 10.1016/j.semcdb.2021.05.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 05/14/2021] [Indexed: 02/06/2023]
Abstract
The advent of induced pluripotent stem cells (iPSCs) and identification of transcription factors for cardiac reprogramming have raised hope to cure heart disease, the leading cause of death in the world. Our knowledge in heart development and molecular barriers of cardiac reprogramming is advancing, but many hurdles are yet to be overcome for clinical translation. Importantly, we lack a full understanding of molecular mechanisms governing cell fate conversion toward cardiomyocytes. In this review, we will discuss the role of metabolism in directed differentiation versus trans-differentiation of cardiomyocytes. Cardiomyocytes exhibit a unique metabolic feature distinct from PSCs and cardiac fibroblasts, and there are multiple overlapping molecular mechanisms underlying metabolic reprogramming during cardiomyogenesis. We will discuss key metabolic changes occurring during cardiomyocytes differentiation from PSCs and cardiac fibroblasts, and the potential role of metabolic reprogramming in the enhancement strategies for cardiomyogenesis. Only when such details are discovered will more effective strategies to enhance the de novo production of cardiomyocytes be possible.
Collapse
|
37
|
Bertuccio SN, Leardini D, Messelodi D, Anselmi L, Manente F, Ragni F, Serravalle S, Masetti R, Pession A. Are Induced Pluripotent Stem Cells a Step towards Modeling Pediatric Leukemias? Cells 2022; 11:cells11030476. [PMID: 35159287 PMCID: PMC8833985 DOI: 10.3390/cells11030476] [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: 12/29/2021] [Revised: 01/26/2022] [Accepted: 01/27/2022] [Indexed: 02/04/2023] Open
Abstract
Despite enormous improvements in pre-clinical and clinical research, acute leukemia still represents an open challenge for pediatric hematologists; both for a significant relapse rate and for long term therapy-related sequelae. In this context, the use of an innovative technology, such as induced pluripotent stem cells (iPSCs), allows to finely reproduce the primary features of the malignancy and can be exploited as a model to study the onset and development of leukemia in vitro. The aim of this review is to explore the recent literature describing iPSCs as a key tool to study different types of hematological malignancies, comprising acute myeloid leukemia, non-down syndrome acute megakaryoblastic leukemia, B cell acute lymphoblastic leukemia, and juvenile myelomonocytic leukemia. This model demonstrates a positive impact on pediatric hematological diseases, especially in those affecting infants whose onsets is found in fetal hematopoiesis. This evidence highlights the importance of achieving an in vitro representation of the human embryonic hematopoietic development and timing-specific modifications, either genetic or epigenetic. Moreover, further insights into clonal evolution studies shed light in the way of a new precision medicine era, where patient-oriented decisions and therapies could further improve the outcome of pediatric cases. Nonetheless, we will also discuss here the difficulties and limitations of this model.
Collapse
Affiliation(s)
- Salvatore Nicola Bertuccio
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (S.N.B.); (F.M.); (F.R.); (R.M.)
| | - Davide Leardini
- Specialty School of Pediatrics, University of Bologna, 40138 Bologna, Italy;
- Pediatric Oncology and Hematology Unit “Lalla Seràgnoli,” Pediatric Unit, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Daria Messelodi
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (S.N.B.); (F.M.); (F.R.); (R.M.)
- Correspondence: (D.M.); (L.A.)
| | - Laura Anselmi
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (S.N.B.); (F.M.); (F.R.); (R.M.)
- Correspondence: (D.M.); (L.A.)
| | - Francesca Manente
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (S.N.B.); (F.M.); (F.R.); (R.M.)
| | - Federico Ragni
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (S.N.B.); (F.M.); (F.R.); (R.M.)
| | - Salvatore Serravalle
- Pediatric Oncology and Hematology Unit “Lalla Seràgnoli,” Pediatric Unit, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Riccardo Masetti
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (S.N.B.); (F.M.); (F.R.); (R.M.)
- Pediatric Oncology and Hematology Unit “Lalla Seràgnoli,” Pediatric Unit, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Andrea Pession
- Division of Pediatrics, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| |
Collapse
|
38
|
Pu WT. Experimental models of Barth syndrome. J Inherit Metab Dis 2022; 45:72-81. [PMID: 34370877 PMCID: PMC8814986 DOI: 10.1002/jimd.12423] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/01/2021] [Accepted: 08/05/2021] [Indexed: 01/03/2023]
Abstract
Mutation of the gene Tafazzin (TAZ) causes Barth syndrome, an X-linked disorder characterized by cardiomyopathy, skeletal muscle weakness, and neutropenia. TAZ is an acyltransferase that catalyzes the remodeling of cardiolipin, the signature phospholipid of the inner mitochondrial membrane. Here, we review the major model systems that have been established to study the role of cardiolipin remodeling in mitochondrial function and the pathogenesis of Barth syndrome. We summarize key features of each model and provide examples of how each has contributed to advance our understanding of TAZ function and Barth syndrome pathophysiology.
Collapse
Affiliation(s)
- William T. Pu
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave., Boston, MA 02115
- Harvard Stem Cell Institute, Harvard University, 7 Divinity Ave, Cambridge, MA 02138
- correspondence:
| |
Collapse
|
39
|
Pérez-Sosa C, Sanluis-Verdes A, Waisman A, Lombardi A, Rosero G, Greca AL, Bhansali S, Bourguignon N, Luzzani C, Pérez MS, Miriuka S, Lerner B. Single cell transfection of human-induced pluripotent stem cells using a droplet-based microfluidic system. ROYAL SOCIETY OPEN SCIENCE 2022; 9:211510. [PMID: 35242349 PMCID: PMC8753139 DOI: 10.1098/rsos.211510] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 12/03/2021] [Indexed: 06/07/2023]
Abstract
Microfluidic tools have recently made possible many advances in biological and biomedical research. Research in fields such as physics, engineering, chemistry and biology have combined to produce innovation in microfluidics which has positively impacted diverse areas such as nucleotide sequencing, functional genomics, single-cell studies, single molecules assays and biomedical diagnostics. Among these areas, regenerative medicine and stem cells have benefited from microfluidics since these tools have had a profound impact on their applications. In this study, we present a high-performance droplet-based system for transfecting individual human-induced pluripotent stem cells. We will demonstrate that this system has great efficiency in single cells and captured droplets, like other microfluidic methods but with lower cost. Moreover, this microfluidic approach can be associated with the PiggyBac transposase-based system to increase its transfection efficiency. Our results provide a starting point for subsequent applications in more complex transfection systems, single-cell differentiation interactions, cell subpopulations and cell therapy, among other potential applications.
Collapse
Affiliation(s)
- Camilo Pérez-Sosa
- National Technological University (UTN), IREN Center, Buenos Aires, Argentina
- National Scientific and Technical Research Council (CONICET) - Foundation for the Fight Against Childhood Neurological Diseases, (LIAN-CONICET-FLENI), FLENI Escobar Headquarters, Route 9 Km 53, 1625, Belén de Escobar, Buenos Aires, Argentina
| | | | - Ariel Waisman
- National Scientific and Technical Research Council (CONICET) - Foundation for the Fight Against Childhood Neurological Diseases, (LIAN-CONICET-FLENI), FLENI Escobar Headquarters, Route 9 Km 53, 1625, Belén de Escobar, Buenos Aires, Argentina
| | - Antonella Lombardi
- National Scientific and Technical Research Council (CONICET) - Foundation for the Fight Against Childhood Neurological Diseases, (LIAN-CONICET-FLENI), FLENI Escobar Headquarters, Route 9 Km 53, 1625, Belén de Escobar, Buenos Aires, Argentina
| | - Gustavo Rosero
- National Technological University (UTN), IREN Center, Buenos Aires, Argentina
| | - Alejandro La Greca
- National Scientific and Technical Research Council (CONICET) - Foundation for the Fight Against Childhood Neurological Diseases, (LIAN-CONICET-FLENI), FLENI Escobar Headquarters, Route 9 Km 53, 1625, Belén de Escobar, Buenos Aires, Argentina
| | - Shekhar Bhansali
- Department of Electrical and Computer Engineering, Florida International University, Miami, FL 33174, USA
| | - Natalia Bourguignon
- National Technological University (UTN), IREN Center, Buenos Aires, Argentina
- Department of Electrical and Computer Engineering, Florida International University, Miami, FL 33174, USA
| | - Carlos Luzzani
- National Scientific and Technical Research Council (CONICET) - Foundation for the Fight Against Childhood Neurological Diseases, (LIAN-CONICET-FLENI), FLENI Escobar Headquarters, Route 9 Km 53, 1625, Belén de Escobar, Buenos Aires, Argentina
| | - Maximiliano. S. Pérez
- University of Buenos Aires (UBA), Institute of Biomedical Engineering, Paseo Colon 850, C1428EGA Buenos Aires, Argentina
- Department of Electrical and Computer Engineering, Florida International University, Miami, FL 33174, USA
| | - Santiago Miriuka
- National Scientific and Technical Research Council (CONICET) - Foundation for the Fight Against Childhood Neurological Diseases, (LIAN-CONICET-FLENI), FLENI Escobar Headquarters, Route 9 Km 53, 1625, Belén de Escobar, Buenos Aires, Argentina
| | - Betiana Lerner
- National Technological University (UTN), IREN Center, Buenos Aires, Argentina
- Department of Electrical and Computer Engineering, Florida International University, Miami, FL 33174, USA
| |
Collapse
|
40
|
Pervaiz I, Al-Ahmad AJ. In Vitro Models of the Human Blood-Brain Barrier Utilising Human Induced Pluripotent Stem Cells: Opportunities and Challenges. Methods Mol Biol 2022; 2492:53-72. [PMID: 35733038 DOI: 10.1007/978-1-0716-2289-6_3] [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] [Indexed: 06/15/2023]
Abstract
The blood-brain barrier (BBB) is a component of the neurovascular unit formed by specialized brain microvascular endothelial cells surrounded by astrocytes end-feet processes, pericytes, and a basement membrane. The BBB plays an important role in the maintenance of brain homeostasis and has seen a growing involvement in the pathophysiology of various neurological diseases. On the other hand, the presence of such a barrier remains an important challenge for drug delivery to treat such illnesses.Since the pioneering work describing the isolation and cultivation of primary brain microvascular cells about 50 years ago until now, the development of an in vitro model of the BBB that is scalable, capable to form tight monolayers, and predictive of drug permeability in vivo remained extremely challenging.The recent description of the use of induced pluripotent stem cells (iPSCs) as a modeling tool for neurological diseases raised momentum into the use of such cells to develop new in vitro models of the BBB. This chapter will provide an exhaustive description of the use of iPSCs as a source of cells for modeling the BBB in vitro, describe the advantages and limitations of such model, as well as describe their prospective use for disease modeling and drug permeability screening platforms.
Collapse
Affiliation(s)
- Iqra Pervaiz
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Abraham J Al-Ahmad
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA.
| |
Collapse
|
41
|
Bone Morphogenetic Protein 4 (BMP4) Enhances the Differentiation of Human Induced Pluripotent Stem Cells into Limbal Progenitor Cells. Curr Issues Mol Biol 2021; 43:2124-2134. [PMID: 34940121 PMCID: PMC8929048 DOI: 10.3390/cimb43030147] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/14/2021] [Accepted: 11/23/2021] [Indexed: 12/21/2022] Open
Abstract
Corneal epithelium maintains visual acuity and is regenerated by the proliferation and differentiation of limbal progenitor cells. Transplantation of human limbal progenitor cells could restore the integrity and functionality of the corneal surface in patients with limbal stem cell deficiency. However, multiple protocols are employed to differentiate human induced pluripotent stem (iPS) cells into corneal epithelium or limbal progenitor cells. The aim of this study was to optimize a protocol that uses bone morphogenetic protein 4 (BMP4) and limbal cell-specific medium. Human dermal fibroblast-derived iPS cells were differentiated into limbal progenitor cells using limbal cell-specific (PI) medium and varying doses (1, 10, and 50 ng/mL) and durations (1, 3, and 10 days) of BMP4 treatment. Differentiated human iPS cells were analyzed by real-time polymerase chain reaction (RT-PCR), Western blotting, and immunocytochemical studies at 2 or 4 weeks after BMP4 treatment. Culturing human dermal fibroblast-derived iPS cells in limbal cell-specific medium and BMP4 gave rise to limbal progenitor and corneal epithelial-like cells. The optimal protocol of 10 ng/mL and three days of BMP4 treatment elicited significantly higher limbal progenitor marker (ABCG2, ∆Np63α) expression and less corneal epithelial cell marker (CK3, CK12) expression than the other combinations of BMP4 dose and duration. In conclusion, this study identified a successful reprogramming strategy to induce limbal progenitor cells from human iPS cells using limbal cell-specific medium and BMP4. Additionally, our experiments indicate that the optimal BMP4 dose and duration favor limbal progenitor cell differentiation over corneal epithelial cells and maintain the phenotype of limbal stem cells. These findings contribute to the development of therapies for limbal stem cell deficiency disorders.
Collapse
|
42
|
Directed Differentiation of Human Pluripotent Stem Cells toward Skeletal Myogenic Progenitors and Their Purification Using Surface Markers. Cells 2021; 10:cells10102746. [PMID: 34685726 PMCID: PMC8534564 DOI: 10.3390/cells10102746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/06/2021] [Accepted: 10/09/2021] [Indexed: 11/17/2022] Open
Abstract
Advancements in reprogramming somatic cells into induced pluripotent stem cells (iPSCs) have provided a strong framework for in vitro disease modeling, gene correction and stem cell-based regenerative medicine. In cases of skeletal muscle disorders, iPSCs can be used for the generation of skeletal muscle progenitors to study disease mechanisms, or implementation for the treatment of muscle disorders. We have recently developed an improved directed differentiation method for the derivation of skeletal myogenic progenitors from hiPSCs. This method allows for a short-term (2 weeks) and efficient skeletal myogenic induction (45-65% of the cells) in human pluripotent stem cells (ESCs/iPSCs) using small molecules to induce mesoderm and subsequently myotomal progenitors, without the need for any gene integration or modification. After initial differentiation, skeletal myogenic progenitors can be purified from unwanted cells using surface markers (CD10+CD24-). These myogenic progenitors have been extensively characterized using in vitro gene expression/differentiation profiling as well as in vivo engraftment studies in dystrophic (mdx) and muscle injury (VML) rodent models and have been proven to be able to engraft and form mature myofibers as well as seeding muscle stem cells. The current protocol describes a detailed, step-by-step guide for this method and outlines important experimental details and troubleshooting points for its application in any human pluripotent stem cells.
Collapse
|
43
|
Disease Modeling of Mitochondrial Cardiomyopathy Using Patient-Specific Induced Pluripotent Stem Cells. BIOLOGY 2021; 10:biology10100981. [PMID: 34681080 PMCID: PMC8533352 DOI: 10.3390/biology10100981] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/25/2021] [Accepted: 09/26/2021] [Indexed: 12/15/2022]
Abstract
Mitochondrial cardiomyopathy (MCM) is characterized as an oxidative phosphorylation disorder of the heart. More than 100 genetic variants in nuclear or mitochondrial DNA have been associated with MCM. However, the underlying molecular mechanisms linking genetic variants to MCM are not fully understood due to the lack of appropriate cellular and animal models. Patient-specific induced pluripotent stem cell (iPSC)-derived cardiomyocytes (iPSC-CMs) provide an attractive experimental platform for modeling cardiovascular diseases and predicting drug efficacy to such diseases. Here we introduce the pathological and therapeutic studies of MCM using iPSC-CMs and discuss the questions and latest strategies for research using iPSC-CMs.
Collapse
|
44
|
Meiser I, Majer J, Katsen-Globa A, Schulz A, Schmidt K, Stracke F, Koutsouraki E, Witt G, Keminer O, Pless O, Gardner J, Claussen C, Gribbon P, Neubauer JC, Zimmermann H. Droplet-based vitrification of adherent human induced pluripotent stem cells on alginate microcarrier influenced by adhesion time and matrix elasticity. Cryobiology 2021; 103:57-69. [PMID: 34582849 DOI: 10.1016/j.cryobiol.2021.09.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/22/2021] [Accepted: 09/23/2021] [Indexed: 10/20/2022]
Abstract
The gold standard in cryopreservation is still conventional slow freezing of single cells or small aggregates in suspension, although major cell loss and limitation to non-specialised cell types in stem cell technology are known drawbacks. The requirement for rapidly available therapeutic and diagnostic cell types is increasing constantly. In the case of human induced pluripotent stem cells (hiPSCs) or their derivates, more sophisticated cryopreservation protocols are needed to address this demand. These should allow a preservation in their physiological, adherent state, an efficient re-cultivation and upscaling upon thawing towards high-throughput applications in cell therapies or disease modelling in drug discovery. Here, we present a novel vitrification-based method for adherent hiPSCs, designed for automated handling by microfluidic approaches and with ready-to-use potential e.g. in suspension-based bioreactors after thawing. Modifiable alginate microcarriers serve as a growth surface for adherent hiPSCs that were cultured in a suspension-based bioreactor and subsequently cryopreserved via droplet-based vitrification in comparison to conventional slow freezing. Soft (0.35%) versus stiff (0.65%) alginate microcarriers in concert with adhesion time variation have been examined. Findings revealed specific optimal conditions leading to an adhesion time and growth surface (matrix) elasticity dependent hypothesis on cryo-induced damaging regimes for adherent cell types. Deviations from the found optimum parameters give rise to membrane ruptures assessed via SEM and major cell loss after adherent vitrification. Applying the optimal conditions, droplet-based vitrification was superior to conventional slow freezing. A decreased microcarrier stiffness was found to outperform stiffer material regarding cell recovery, whereas the stemness characteristics of rewarmed hiPSCs were preserved.
Collapse
Affiliation(s)
- Ina Meiser
- Fraunhofer Institute for Biomedical Engineering IBMT, 66280, Sulzbach, Saar, Germany.
| | - Julia Majer
- Fraunhofer Institute for Biomedical Engineering IBMT, 66280, Sulzbach, Saar, Germany
| | - Alisa Katsen-Globa
- Fraunhofer Institute for Biomedical Engineering IBMT, 66280, Sulzbach, Saar, Germany
| | - André Schulz
- Fraunhofer Institute for Biomedical Engineering IBMT, 66280, Sulzbach, Saar, Germany
| | - Katharina Schmidt
- Fraunhofer Institute for Biomedical Engineering IBMT, 66280, Sulzbach, Saar, Germany
| | - Frank Stracke
- Fraunhofer Institute for Biomedical Engineering IBMT, 66280, Sulzbach, Saar, Germany
| | | | - Gesa Witt
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, ScreeningPort, 22525, Hamburg, Germany
| | - Oliver Keminer
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, ScreeningPort, 22525, Hamburg, Germany
| | - Ole Pless
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, ScreeningPort, 22525, Hamburg, Germany
| | - John Gardner
- Censo Biotechnologies Ltd, Roslin Midlothian, EH25 9RG, United Kingdom
| | - Carsten Claussen
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, ScreeningPort, 22525, Hamburg, Germany
| | - Philip Gribbon
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, ScreeningPort, 22525, Hamburg, Germany
| | - Julia C Neubauer
- Fraunhofer Institute for Biomedical Engineering IBMT, 66280, Sulzbach, Saar, Germany; Fraunhofer Project Centre for Stem Cell Process Engineering, 97081, Würzburg, Germany
| | - Heiko Zimmermann
- Fraunhofer Institute for Biomedical Engineering IBMT, 66280, Sulzbach, Saar, Germany; Censo Biotechnologies Ltd, Roslin Midlothian, EH25 9RG, United Kingdom; Faculty of Marine Science, Universidad Católica Del Norte, 1781421, Coquimbo, Chile; Chair for Molecular and Cellular Biotechnology / Nanotechnology, Saarland University, 66123, Saarbrücken, Germany
| |
Collapse
|
45
|
Shcheglovitov A, Peterson RT. Screening Platforms for Genetic Epilepsies-Zebrafish, iPSC-Derived Neurons, and Organoids. Neurotherapeutics 2021; 18:1478-1489. [PMID: 34595731 PMCID: PMC8608971 DOI: 10.1007/s13311-021-01115-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2021] [Indexed: 02/04/2023] Open
Abstract
Recent advances in molecular and cellular engineering, such as human cell reprogramming, genome editing, and patient-specific organoids, have provided unprecedented opportunities for investigating human disorders in both animals and human-based models at an improved pace and precision. This progress will inevitably lead to the development of innovative drug-screening platforms and new patient-specific therapeutics. In this review, we discuss recent advances that have been made using zebrafish and human-induced pluripotent stem cell (iPSC)-derived neurons and organoids for modeling genetic epilepsies. We also provide our prospective on how these models can potentially be combined to build new screening platforms for antiseizure and antiepileptogenic drug discovery that harness the robustness and tractability of zebrafish models as well as the patient-specific genetics and biology of iPSC-derived neurons and organoids.
Collapse
|
46
|
Tenreiro MF, Louro AF, Alves PM, Serra M. Next generation of heart regenerative therapies: progress and promise of cardiac tissue engineering. NPJ Regen Med 2021; 6:30. [PMID: 34075050 PMCID: PMC8169890 DOI: 10.1038/s41536-021-00140-4] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 05/10/2021] [Indexed: 02/04/2023] Open
Abstract
The adult heart is a vital and highly specialized organ of the human body, with limited capability of self-repair and regeneration in case of injury or disease. Engineering biomimetic cardiac tissue to regenerate the heart has been an ambition in the field of tissue engineering, tracing back to the 1990s. Increased understanding of human stem cell biology and advances in process engineering have provided an unlimited source of cells, particularly cardiomyocytes, for the development of functional cardiac muscle, even though pluripotent stem cell-derived cardiomyocytes poorly resemble those of the adult heart. This review outlines key biology-inspired strategies reported to improve cardiomyocyte maturation features and current biofabrication approaches developed to engineer clinically relevant cardiac tissues. It also highlights the potential use of this technology in drug discovery science and disease modeling as well as the current efforts to translate it into effective therapies that improve heart function and promote regeneration.
Collapse
Affiliation(s)
- Miguel F Tenreiro
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal
- Instituto de Tecnologia Química e Biologica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Ana F Louro
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal
- Instituto de Tecnologia Química e Biologica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Paula M Alves
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal
- Instituto de Tecnologia Química e Biologica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Margarida Serra
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal.
- Instituto de Tecnologia Química e Biologica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal.
| |
Collapse
|
47
|
Bang S, Lee S, Choi N, Kim HN. Emerging Brain-Pathophysiology-Mimetic Platforms for Studying Neurodegenerative Diseases: Brain Organoids and Brains-on-a-Chip. Adv Healthc Mater 2021; 10:e2002119. [PMID: 34028201 DOI: 10.1002/adhm.202002119] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 03/25/2021] [Indexed: 12/13/2022]
Abstract
Neurodegenerative diseases are a group of disorders characterized by progressive degeneration of the structural and functional integrity of the central and peripheral nervous systems. Millions of people suffer from degenerative brain diseases worldwide, and the mortality continues to increase every year, causing a growing demand for knowledge of the underlying mechanisms and development of therapeutic targets. Conventional 2D-based cell culture platforms and animal models cannot fully recapitulate the pathophysiology, and this has limited the capability for estimating drug efficacy. Recently, engineered platforms, including brain organoids and brain-on-a-chip, have emerged. They mimic the physiology of brain tissue and reflect the fundamental pathophysiological signatures of neurodegenerative diseases, such as the accumulation of neurotoxic proteins, structural abnormalities, and functional loss. In this paper, recent advances in brain-mimetic platforms and their potential for modeling features of neurodegenerative diseases in vitro are reviewed. The development of a physiologically relevant model should help overcome unresolved neurodegenerative diseases.
Collapse
Affiliation(s)
- Seokyoung Bang
- Brain Science Institute Korea Institute of Science and Technology (KIST) Seoul 02792 Republic of Korea
| | - Songhyun Lee
- Department of Medical Engineering Yonsei University College of Medicine Seoul 03722 Republic of Korea
| | - Nakwon Choi
- Brain Science Institute Korea Institute of Science and Technology (KIST) Seoul 02792 Republic of Korea
- KU‐KIST Graduate School of Converging Science and Technology Korea University Seoul 02841 Republic of Korea
| | - Hong Nam Kim
- Brain Science Institute Korea Institute of Science and Technology (KIST) Seoul 02792 Republic of Korea
- Division of Bio‐Medical Science & Technology KIST School Korea University of Science and Technology (UST) Seoul 02792 Republic of Korea
| |
Collapse
|
48
|
De la Vega L, Abelseth L, Sharma R, Triviño-Paredes J, Restan M, Willerth SM. 3D Bioprinting Human‐Induced Pluripotent Stem Cells and Drug‐Releasing Microspheres to Produce Responsive Neural Tissues. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202000077] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Laura De la Vega
- Department of Mechanical Engineering University of Victoria Victoria V8W 2Y2 Canada
| | - Laila Abelseth
- Biomedical Engineering Program University of Victoria Victoria V8W 2Y2 Canada
| | - Ruchi Sharma
- Department of Mechanical Engineering University of Victoria Victoria V8W 2Y2 Canada
| | | | - Milena Restan
- Biomedical Engineering Program University of Victoria Victoria V8W 2Y2 Canada
| | - Stephanie M. Willerth
- Department of Mechanical Engineering University of Victoria Victoria V8W 2Y2 Canada
- Biomedical Engineering Program University of Victoria Victoria V8W 2Y2 Canada
- Division of Medical Sciences University of Victoria Victoria V8W 2Y2 Canada
| |
Collapse
|
49
|
Joshi K, Cameron F, Tiwari S, Mannering SI, Elefanty AG, Stanley EG. Modeling Type 1 Diabetes Using Pluripotent Stem Cell Technology. Front Endocrinol (Lausanne) 2021; 12:635662. [PMID: 33868170 PMCID: PMC8047192 DOI: 10.3389/fendo.2021.635662] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 03/03/2021] [Indexed: 12/26/2022] Open
Abstract
Induced pluripotent stem cell (iPSC) technology is increasingly being used to create in vitro models of monogenic human disorders. This is possible because, by and large, the phenotypic consequences of such genetic variants are often confined to a specific and known cell type, and the genetic variants themselves can be clearly identified and controlled for using a standardized genetic background. In contrast, complex conditions such as autoimmune Type 1 diabetes (T1D) have a polygenic inheritance and are subject to diverse environmental influences. Moreover, the potential cell types thought to contribute to disease progression are many and varied. Furthermore, as HLA matching is critical for cell-cell interactions in disease pathogenesis, any model that seeks to test the involvement of particular cell types must take this restriction into account. As such, creation of an in vitro model of T1D will require a system that is cognizant of genetic background and enables the interaction of cells representing multiple lineages to be examined in the context of the relevant environmental disease triggers. In addition, as many of the lineages critical to the development of T1D cannot be easily generated from iPSCs, such models will likely require combinations of cell types derived from in vitro and in vivo sources. In this review we imagine what an ideal in vitro model of T1D might look like and discuss how the required elements could be feasibly assembled using existing technologies. We also examine recent advances towards this goal and discuss potential uses of this technology in contributing to our understanding of the mechanisms underlying this autoimmune condition.
Collapse
Affiliation(s)
- Kriti Joshi
- Department of Endocrinology and Metabolism, All India Institute of Medical Sciences Rishikesh, Uttarakhand, India
- Department of Molecular Medicine & Biotechnology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India
- Department of Cell Biology, Murdoch Children’s Research Institute, Parkville, Vic, Australia
| | - Fergus Cameron
- Department of Cell Biology, Murdoch Children’s Research Institute, Parkville, Vic, Australia
- Department of Endocrinology and Diabetes, The Royal Children’s Hospital, Parkville, Vic, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Vic, Australia
| | - Swasti Tiwari
- Department of Molecular Medicine & Biotechnology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India
| | - Stuart I. Mannering
- Immunology and Diabetes Unit, St. Vincent’s Institute of Medical Research, Fitzroy, Vic, Australia
| | - Andrew G. Elefanty
- Department of Cell Biology, Murdoch Children’s Research Institute, Parkville, Vic, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Vic, Australia
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Vic, Australia
| | - Edouard G. Stanley
- Department of Cell Biology, Murdoch Children’s Research Institute, Parkville, Vic, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Vic, Australia
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Vic, Australia
| |
Collapse
|
50
|
Neurospheres: a potential in vitro model for the study of central nervous system disorders. Mol Biol Rep 2021; 48:3649-3663. [PMID: 33765252 DOI: 10.1007/s11033-021-06301-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 03/18/2021] [Indexed: 02/08/2023]
Abstract
Neurogenesis was believed to end after the period of embryonic development. However, the possibility of obtaining an expressive number of cells with functional neuronal characteristics implied a great advance in experimental research. New techniques have emerged to demonstrate that the birth of new neurons continues to occur in the adult brain. Two main rich sources of these cells are the subventricular zone (SVZ) and the subgranular zone of the hippocampal dentate gyrus (SGZ) where adult neural stem cells (aNSCs) have the ability to proliferate and differentiate into mature cell lines. The cultivation of neurospheres is a method to isolate, maintain and expand neural stem cells (NSCs) and has been used extensively by several research groups to analyze the biological properties of NSCs and their potential use in injured brains from animal models. Throughout this review, we highlight the areas where this type of cell culture has been applied and the advantages and limitations of using this model in experimental studies for the neurological clinical scenario.
Collapse
|