1
|
Rohban R, Martins CP, Esni F. Advanced therapy to cure diabetes: mission impossible is now possible? Front Cell Dev Biol 2024; 12:1484859. [PMID: 39629270 PMCID: PMC11611888 DOI: 10.3389/fcell.2024.1484859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024] Open
Abstract
Cell and Gene therapy are referred to as advanced therapies that represent overlapping fields of regenerative medicine. They have similar therapeutic goals such as to modify cellular identity, improve cell function, or fight a disease. These two therapeutic avenues, however, possess major differences. While cell therapy involves introduction of new cells, gene therapy entails introduction or modification of genes. Furthermore, the aim of cell therapy is often to replace, or repair damaged tissue, whereas gene therapy is used typically as a preventive approach. Diabetes mellitus severely affects the quality of life of afflicted individuals and has various side effects including cardiovascular, ophthalmic disorders, and neuropathy while putting enormous economic pressure on both the healthcare system and the patient. In recent years, great effort has been made to develop cutting-edge therapeutic interventions for diabetes treatment, among which cell and gene therapies stand out. This review aims to highlight various cell- and gene-based therapeutic approaches leading to the generation of new insulin-producing cells as a topmost "panacea" for treating diabetes, while deliberately avoiding a detailed molecular description of these approaches. By doing so, we aim to target readers who are new to the field and wish to get a broad helicopter overview of the historical and current trends of cell- and gene-based approaches in β-cell regeneration.
Collapse
Affiliation(s)
- Rokhsareh Rohban
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Christina P. Martins
- Department of Surgery, Division of Pediatric General and Thoracic Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Farzad Esni
- Department of Surgery, Division of Pediatric General and Thoracic Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA, United States
- UPMC Hillman Cancer Center, Pittsburgh, PA, United States
- McGowan Institute for regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
2
|
Mojares E, Nadal C, Hayler D, Kanso H, Chrysanthou A, Neri Cruz CE, Gautrot JE. Strong Elastic Protein Nanosheets Enable the Culture and Differentiation of Induced Pluripotent Stem Cells on Microdroplets. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2406333. [PMID: 39036832 DOI: 10.1002/adma.202406333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 06/30/2024] [Indexed: 07/23/2024]
Abstract
Advances in stem cell technologies, revolutionizing regenerative therapies and advanced in vitro testing, require novel cell manufacturing pipelines able to cope with scale up and parallelization. Microdroplet technologies, which have transformed single cell sequencing and other cell-based assays, are attractive in this context, but the inherent soft mechanics of liquid-liquid interfaces is typically thought to be incompatible with the expansion of induced pluripotent stem cells (iPSCs), and their differentiation. In this work, the design of protein nanosheets stabilizing liquid-liquid interfaces and enabling the adhesion, expansion and retention of stemness by iPSCs is reported. Microdroplet microfluidic chips are used to control the formulation of droplets with defined dimensions and size distributions. The resulting emulsions sustain high expansion rates, with excellent retention of stem cell marker expression. iPSCs cultured in such conditions retain the capacity to differentiate into cardiomyocytes. This work provides clear evidence that local nanoscale mechanics, associated with interfacial viscoelasticity, provides strong cues able to regulate and maintain pluripotency, as well as to support commitment in defined differentiation conditions. Microdroplet technologies appear as attractive candidates to transform cell manufacturing pipelines, bypassing significant hurdles paused by solid substrates and microcarriers.
Collapse
Affiliation(s)
- Elijah Mojares
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London, E1 4NS, UK
| | - Clemence Nadal
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London, E1 4NS, UK
| | - Daniel Hayler
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London, E1 4NS, UK
| | - Hassan Kanso
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London, E1 4NS, UK
| | - Alexandra Chrysanthou
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London, E1 4NS, UK
| | - Carlos E Neri Cruz
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London, E1 4NS, UK
| | - Julien E Gautrot
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London, E1 4NS, UK
| |
Collapse
|
3
|
Liang S, Zhou J, Yu X, Lu S, Liu R. Neuronal conversion from glia to replenish the lost neurons. Neural Regen Res 2024; 19:1446-1453. [PMID: 38051886 PMCID: PMC10883502 DOI: 10.4103/1673-5374.386400] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 08/16/2023] [Indexed: 12/07/2023] Open
Abstract
ABSTRACT Neuronal injury, aging, and cerebrovascular and neurodegenerative diseases such as cerebral infarction, Alzheimer's disease, Parkinson's disease, frontotemporal dementia, amyotrophic lateral sclerosis, and Huntington's disease are characterized by significant neuronal loss. Unfortunately, the neurons of most mammals including humans do not possess the ability to self-regenerate. Replenishment of lost neurons becomes an appealing therapeutic strategy to reverse the disease phenotype. Transplantation of pluripotent neural stem cells can supplement the missing neurons in the brain, but it carries the risk of causing gene mutation, tumorigenesis, severe inflammation, and obstructive hydrocephalus induced by brain edema. Conversion of neural or non-neural lineage cells into functional neurons is a promising strategy for the diseases involving neuron loss, which may overcome the above-mentioned disadvantages of neural stem cell therapy. Thus far, many strategies to transform astrocytes, fibroblasts, microglia, Müller glia, NG2 cells, and other glial cells to mature and functional neurons, or for the conversion between neuronal subtypes have been developed through the regulation of transcription factors, polypyrimidine tract binding protein 1 (PTBP1), and small chemical molecules or are based on a combination of several factors and the location in the central nervous system. However, some recent papers did not obtain expected results, and discrepancies exist. Therefore, in this review, we discuss the history of neuronal transdifferentiation, summarize the strategies for neuronal replenishment and conversion from glia, especially astrocytes, and point out that biosafety, new strategies, and the accurate origin of the truly converted neurons in vivo should be focused upon in future studies. It also arises the attention of replenishing the lost neurons from glia by gene therapies such as up-regulation of some transcription factors or down-regulation of PTBP1 or drug interference therapies.
Collapse
Affiliation(s)
- Shiyu Liang
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Jing Zhou
- Department of Geriatric Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Xiaolin Yu
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| | - Shuai Lu
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| | - Ruitian Liu
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
4
|
Dhanjal DS, Singh R, Sharma V, Nepovimova E, Adam V, Kuca K, Chopra C. Advances in Genetic Reprogramming: Prospects from Developmental Biology to Regenerative Medicine. Curr Med Chem 2024; 31:1646-1690. [PMID: 37138422 DOI: 10.2174/0929867330666230503144619] [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: 11/12/2022] [Revised: 03/13/2023] [Accepted: 03/16/2023] [Indexed: 05/05/2023]
Abstract
The foundations of cell reprogramming were laid by Yamanaka and co-workers, who showed that somatic cells can be reprogrammed into pluripotent cells (induced pluripotency). Since this discovery, the field of regenerative medicine has seen advancements. For example, because they can differentiate into multiple cell types, pluripotent stem cells are considered vital components in regenerative medicine aimed at the functional restoration of damaged tissue. Despite years of research, both replacement and restoration of failed organs/ tissues have remained elusive scientific feats. However, with the inception of cell engineering and nuclear reprogramming, useful solutions have been identified to counter the need for compatible and sustainable organs. By combining the science underlying genetic engineering and nuclear reprogramming with regenerative medicine, scientists have engineered cells to make gene and stem cell therapies applicable and effective. These approaches have enabled the targeting of various pathways to reprogramme cells, i.e., make them behave in beneficial ways in a patient-specific manner. Technological advancements have clearly supported the concept and realization of regenerative medicine. Genetic engineering is used for tissue engineering and nuclear reprogramming and has led to advances in regenerative medicine. Targeted therapies and replacement of traumatized , damaged, or aged organs can be realized through genetic engineering. Furthermore, the success of these therapies has been validated through thousands of clinical trials. Scientists are currently evaluating induced tissue-specific stem cells (iTSCs), which may lead to tumour-free applications of pluripotency induction. In this review, we present state-of-the-art genetic engineering that has been used in regenerative medicine. We also focus on ways that genetic engineering and nuclear reprogramming have transformed regenerative medicine and have become unique therapeutic niches.
Collapse
Affiliation(s)
- Daljeet Singh Dhanjal
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| | - Reena Singh
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| | - Varun Sharma
- Head of Bioinformatic Division, NMC Genetics India Pvt. Ltd., Gurugram, India
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, 50003, Czech Republic
| | - Vojtech Adam
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, Brno, CZ 613 00, Czech Republic
- Central European Institute of Technology, Brno University of Technology, Purkynova 123, Brno, CZ-612 00, Czech Republic
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, 50003, Czech Republic
- Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, 50005, Czech Republic
| | - Chirag Chopra
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| |
Collapse
|
5
|
Matos BMD, Stimamiglio MA, Correa A, Robert AW. Human pluripotent stem cell-derived extracellular vesicles: From now to the future. World J Stem Cells 2023; 15:453-465. [PMID: 37342215 PMCID: PMC10277970 DOI: 10.4252/wjsc.v15.i5.453] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 02/14/2023] [Accepted: 04/13/2023] [Indexed: 05/26/2023] Open
Abstract
Extracellular vesicles (EVs) are nanometric particles that enclose cell-derived bioactive molecules in a lipid bilayer and serve as intercellular communication tools. Accordingly, in various biological contexts, EVs are reported to engage in immune modulation, senescence, and cell proliferation and differentiation. Therefore, EVs could be key elements for potential off-the-shelf cell-free therapy. Little has been studied regarding EVs derived from human pluripotent stem cells (hPSC-EVs), even though hPSCs offer good opportunities for induction of tissue regeneration and unlimited proliferative ability. In this review article, we provide an overview of studies using hPSC-EVs, focusing on identifying the conditions in which the cells are cultivated for the isolation of EVs, how they are characterized, and applications already demonstrated. The topics reported in this article highlight the incipient status of the studies in the field and the significance of hPSC-EVs’ prospective applications as PSC-derived cell-free therapy products.
Collapse
Affiliation(s)
- Bruno Moises de Matos
- Stem Cells Basic Biology Laboratory, Carlos Chagas Institute, Curitiba 81350010, Paraná, Brazil
| | | | - Alejandro Correa
- Stem Cells Basic Biology Laboratory, Carlos Chagas Institute, Curitiba 81350010, Paraná, Brazil
| | - Anny Waloski Robert
- Stem Cells Basic Biology Laboratory, Carlos Chagas Institute, Curitiba 81350010, Paraná, Brazil
| |
Collapse
|
6
|
Desa DE, Qian T, Skala MC. Label-free optical imaging and sensing for quality control of stem cell manufacturing. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2023; 25:100435. [PMID: 37885458 PMCID: PMC10602581 DOI: 10.1016/j.cobme.2022.100435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Human stem cells provide emerging methods for drug screening, disease modeling, and personalized patient therapies. To meet this growing demand for scale-up, stem cell manufacturing methods must be streamlined with continuous monitoring technologies and automated feedback to optimize growth conditions for high production and consistency. Label-free optical imaging and sensing, including multiphoton microscopy, Raman spectroscopy, and low-cost methods such as phase and transmitted light microscopy, can provide rapid, repeatable, and non-invasive monitoring of stem cells throughout cell differentiation and maturation. Machine learning algorithms trained on label-free optical imaging and sensing features could identify viable cells and predict optimal manufacturing conditions. These techniques have the potential to streamline stem cell manufacturing and accelerate their use in regenerative medicine.
Collapse
Affiliation(s)
- Danielle E Desa
- Morgridge Institute for Research, 330 N. Orchard St., Madison, WI 53715, United States
| | - Tongcheng Qian
- Morgridge Institute for Research, 330 N. Orchard St., Madison, WI 53715, United States
| | - Melissa C Skala
- Morgridge Institute for Research, 330 N. Orchard St., Madison, WI 53715, United States
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1550 Engineering Dr., Madison, WI 53706, United States
| |
Collapse
|
7
|
Karami Z, Moradi S, Eidi A, Soleimani M, Jafarian A. Induced pluripotent stem cells: Generation methods and a new perspective in COVID-19 research. Front Cell Dev Biol 2023; 10:1050856. [PMID: 36733338 PMCID: PMC9887183 DOI: 10.3389/fcell.2022.1050856] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 11/22/2022] [Indexed: 01/18/2023] Open
Abstract
Induced pluripotent stem cells (iPSCs) exhibit an unlimited ability to self-renew and produce various differentiated cell types, thereby creating high hopes for both scientists and patients as a great tool for basic research as well as for regenerative medicine purposes. The availability and safety of iPSCs for therapeutic purposes require safe and highly efficient methods for production of these cells. Different methods have been used to produce iPSCs, each of which has advantages and disadvantages. Studying these methods would be very helpful in developing an easy, safe, and efficient method for the generation of iPSCs. Since iPSCs can be generated from somatic cells, they can be considered as valuable cellular resources available for important research needs and various therapeutic purposes. Coronavirus disease 2019 (COVID-19) is a disease that has endangered numerous human lives worldwide and currently has no definitive cure. Therefore, researchers have been rigorously studying and examining all aspects of COVID-19 and potential treatment modalities and various drugs in order to enable the treatment, control, and prevention of COVID-19. iPSCs have become one of the most attractive and promising tools in this field by providing the ability to study COVID-19 and the effectiveness of drugs on this disease outside the human body. In this study, we discuss the different methods of generation of iPSCs as well as their respective advantages and disadvantages. We also present recent applications of iPSCs in the study and treatment of COVID-19.
Collapse
Affiliation(s)
- Zahra Karami
- 1Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Sharif Moradi
- 2Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Akram Eidi
- 1Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Masoud Soleimani
- 3Hematology and Cell Therapy Department, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran,4Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arefeh Jafarian
- 5Iranian Tissue Bank and Research Center, Tehran University of Medical Sciences, Tehran, Iran,*Correspondence: Arefeh Jafarian,
| |
Collapse
|
8
|
Gu X, Wang J, Jiang X. miR-124- and let-7-Mediated Reprogram of Human Fibroblasts into SST Interneurons. ACS Chem Neurosci 2022; 13:2755-2765. [PMID: 36074953 DOI: 10.1021/acschemneuro.2c00445] [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: 01/20/2023] Open
Abstract
Many neurological disorders stem from defects in or the loss of specific neurons. Dysfunction of γ-aminobutyric acid (GABA)ergic interneurons may cause a variety of neurological and psychiatric disorders such as epilepsy, autism, Alzheimer's disease, and depression. Unlike other types of neurons, which can be generated relatively easily by direct reprogramming, it is difficult to generate GABAergic neurons by traditional methods. Neuronal transdifferentiation of fibroblasts mediated by nongenomic-integrated adenovirus has many advantages, but the efficiency is low, and there is a lack of studies using human cells as the initial materials. In this study, we explored the feasibility of the conversion of human fibroblasts into neurons through adenovirus-mediated gene expression and found that by introducing two microRNAs, miR-124 and let-7, together with several small chemical compounds, they can effectively generate GABAergic neuron-like cells from human neonatal fibroblasts without reverting to a progenitor cell stage. Most of these cells expressed neuronal markers and were all somatostatin (SST)-positive cells. Therefore, our study provides a relatively safe and efficient method to generate SST interneurons.
Collapse
Affiliation(s)
- Xi Gu
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510500, China.,Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350000, China
| | - Junhao Wang
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350000, China
| | - Xiaodan Jiang
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510500, China.,Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou 510500, China
| |
Collapse
|
9
|
Chakraborty AR, Vassilev A, Jaiswal SK, O'Connell CE, Ahrens JF, Mallon BS, Pera MF, DePamphilis ML. Selective elimination of pluripotent stem cells by PIKfyve specific inhibitors. Stem Cell Reports 2022; 17:397-412. [PMID: 35063131 PMCID: PMC8828683 DOI: 10.1016/j.stemcr.2021.12.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/15/2021] [Accepted: 12/16/2021] [Indexed: 01/05/2023] Open
Abstract
Inhibition of PIKfyve phosphoinositide kinase selectively kills autophagy-dependent cancer cells by disrupting lysosome homeostasis. Here, we show that PIKfyve inhibitors can also selectively eliminate pluripotent embryonal carcinoma cells (ECCs), embryonic stem cells, and induced pluripotent stem cells under conditions where differentiated cells remain viable. PIKfyve inhibitors prevented lysosome fission, induced autophagosome accumulation, and reduced cell proliferation in both pluripotent and differentiated cells, but they induced death only in pluripotent cells. The ability of PIKfyve inhibitors to distinguish between pluripotent and differentiated cells was confirmed with xenografts derived from ECCs. Pretreatment of ECCs with the PIKfyve specific inhibitor WX8 suppressed their ability to form teratocarcinomas in mice, and intraperitoneal injections of WX8 into mice harboring teratocarcinoma xenografts selectively eliminated pluripotent cells. Differentiated cells continued to proliferate, but at a reduced rate. These results provide a proof of principle that PIKfyve specific inhibitors can selectively eliminate pluripotent stem cells in vivo as well as in vitro.
Collapse
Affiliation(s)
- Arup R Chakraborty
- National Institute of Child Health & Human Development, National Institutes of Health, Bldg. 6A/3A15, 6 Center Drive, Bethesda, MD 20892-2790, USA
| | - Alex Vassilev
- National Institute of Child Health & Human Development, National Institutes of Health, Bldg. 6A/3A15, 6 Center Drive, Bethesda, MD 20892-2790, USA
| | - Sushil K Jaiswal
- National Institute of Child Health & Human Development, National Institutes of Health, Bldg. 6A/3A15, 6 Center Drive, Bethesda, MD 20892-2790, USA
| | - Constandina E O'Connell
- National Institute of Child Health & Human Development, National Institutes of Health, Bldg. 6A/3A15, 6 Center Drive, Bethesda, MD 20892-2790, USA
| | - John F Ahrens
- National Institute of Child Health & Human Development, National Institutes of Health, Bldg. 6A/3A15, 6 Center Drive, Bethesda, MD 20892-2790, USA
| | - Barbara S Mallon
- NIH Stem Cell Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Melvin L DePamphilis
- National Institute of Child Health & Human Development, National Institutes of Health, Bldg. 6A/3A15, 6 Center Drive, Bethesda, MD 20892-2790, USA.
| |
Collapse
|
10
|
Lynch E, Peek E, Reilly M, FitzGibbons C, Robertson S, Suzuki M. Current Progress in the Creation, Characterization, and Application of Human Stem Cell-derived in Vitro Neuromuscular Junction Models. Stem Cell Rev Rep 2022; 18:768-780. [PMID: 34212303 PMCID: PMC8720113 DOI: 10.1007/s12015-021-10201-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2021] [Indexed: 02/03/2023]
Abstract
Human pluripotent stem cells (PSCs) such as embryonic stem cells (ESCs) or induced pluripotent stem cells (iPSCs) are of great value for studying developmental processes, disease modeling, and drug testing. One area in which the use of human PSCs has become of great interest in recent years is for in vitro models of the neuromuscular junction (NMJ). The NMJ is a synapse at which a motor neuron releases acetylcholine to bind to skeletal muscle and stimulate contraction. Degeneration of the NMJ and subsequent loss of muscle function is a common feature of many neuromuscular diseases such as myasthenia gravis, spinal muscular atrophy, and amyotrophic lateral sclerosis. In order to develop new therapies for patients with neuromuscular diseases, it is essential to understand mechanisms taking place at the NMJ. However, we have limited ability to study the NMJ in living human patients, and animal models are limited by physiological relevance. Therefore, an in vitro model of the NMJ consisting of human cells is of great value. The use of stem cells for in vitro NMJ models is still in progress and requires further optimization in order to yield reliable, reproducible results. The objective of this review is (1) to outline the current progress towards fully PSC-derived in vitro co-culture models of the human NMJ and (2) to discuss future directions and challenges that must be overcome in order to create reproducible fully PSC-derived models that can be used for developmental studies, disease modeling, and drug testing.
Collapse
Affiliation(s)
- Eileen Lynch
- Department of Comparative Biosciences, University of Wisconsin-Madison, Wisconsin, USA
| | - Emma Peek
- Department of Comparative Biosciences, University of Wisconsin-Madison, Wisconsin, USA
| | - Megan Reilly
- Department of Comparative Biosciences, University of Wisconsin-Madison, Wisconsin, USA
| | - Claire FitzGibbons
- Department of Comparative Biosciences, University of Wisconsin-Madison, Wisconsin, USA
| | - Samantha Robertson
- Department of Comparative Biosciences, University of Wisconsin-Madison, Wisconsin, USA
| | - Masatoshi Suzuki
- Department of Comparative Biosciences, University of Wisconsin-Madison, Wisconsin, USA,Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, Wisconsin, USA
| |
Collapse
|
11
|
Huang YJ, Cao J, Lee CY, Wu YM. Umbilical cord blood plasma-derived exosomes as a novel therapy to reverse liver fibrosis. Stem Cell Res Ther 2021; 12:568. [PMID: 34772443 PMCID: PMC8588641 DOI: 10.1186/s13287-021-02641-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 10/28/2021] [Indexed: 02/08/2023] Open
Abstract
Background Cirrhosis is a chronic liver disease whereby scar tissue replaces healthy liver parenchyma, leading to disruption of the liver architecture and hepatic dysfunction. Currently, there is no effective disease-modifying therapy for liver fibrosis. Recently, our group demonstrated that human umbilical cord blood (UCB) plasma possesses therapeutic effects in a rat model of acute liver failure. Methods In the current study, we tested whether exosomes (Exo) existed in UCB plasma and if they produced any antifibrotic benefits in a liver fibrosis model. Results Our results showed that UCB-Exo improved liver function and increased matrix metalloproteinase/tissue inhibitor of metalloproteinase degradation to reduce the degree of fibrosis. Moreover, UCB-Exo were found to suppress hepatic stellate cell (HSC) activity in vitro. These effects were associated with suppression of transforming growth factor-β/inhibitor of DNA binding 1 signaling. Conclusions These results further support that UCB-Exo have antifibrotic effects in mice with liver fibrosis and activated HSCs and may herald a new cell-free antifibrotic therapy.
Collapse
Affiliation(s)
- Yu-Jen Huang
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Jerry Cao
- Department of Surgery, Wollongong Hospital, Loftus Street, Wollongong, NSW, 2500, Australia
| | - Chih-Yuan Lee
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Yao-Ming Wu
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan. .,Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, No. 7, Chung-Shan South Road, Taipei, Taiwan.
| |
Collapse
|
12
|
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
|
13
|
Novel therapies using cell sheets engineered from allogeneic mesenchymal stem/stromal cells. Emerg Top Life Sci 2021; 4:677-689. [PMID: 33231260 PMCID: PMC7939697 DOI: 10.1042/etls20200151] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 10/31/2020] [Accepted: 11/05/2020] [Indexed: 01/05/2023]
Abstract
Mesenchymal stem/stromal cells (MSCs) have long been recognized to help regenerate tissues, by exploiting their intrinsic potentials for differentiation and secretion of therapeutic paracrine factors together with feasibility for cell banking. These unique MSC properties are attractive to provide effective new cell-based therapies for unmet medical needs. Currently, the infusion of suspended MSCs is accepted as a promising therapy to treat systemic inflammatory diseases. However, low cell engraftment/retention in target organs and off-target entrapment using conventional cell infusion must be improved to provide reliable localized disease treatments. Cell sheet technology offers an alternative: three-dimensional (3D) tissue-like structures can be harvested from culture using mild temperature reduction, and transplanted directly onto target tissue sites without suturing, yielding stable cell engraftment and prolonged cell retention in situ without off-target losses. Engineered MSC sheets directly address two major cell therapy strategies based on their therapeutic benefits: (1) tissue replacements based on mult-ilineage differentiation capacities, focusing on cartilage regeneration in this review, and (2) enhancement of tissue recovery via paracrine signaling, employing their various secreted cytokines to promote neovascularization. MSCs also have production benefits as a promising allogeneic cell source by exploiting their reliable proliferative capacity to facilitate expansion and sustainable cell banking for off-the-shelf therapies. This article reviews the advantages of both MSCs as allogeneic cell sources in contrast with autologous cell sources, and allogeneic MSC sheets engineered on thermo-responsive cell dishes as determined in basic studies and clinical achievements, indicating promise to provide robust new cell therapies to future patients.
Collapse
|
14
|
Wang Y, Zhang X, Chen F, Song N, Xie J. In vivo Direct Conversion of Astrocytes to Neurons Maybe a Potential Alternative Strategy for Neurodegenerative Diseases. Front Aging Neurosci 2021; 13:689276. [PMID: 34408642 PMCID: PMC8366583 DOI: 10.3389/fnagi.2021.689276] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/08/2021] [Indexed: 01/06/2023] Open
Abstract
Partly because of extensions in lifespan, the incidence of neurodegenerative diseases is increasing, while there is no effective approach to slow or prevent neuronal degeneration. As we all know, neurons cannot self-regenerate and may not be replaced once being damaged or degenerated in human brain. Astrocytes are widely distributed in the central nervous system (CNS) and proliferate once CNS injury or neurodegeneration occur. Actually, direct reprogramming astrocytes into functional neurons has been attracting more and more attention in recent years. Human astrocytes can be successfully converted into neurons in vitro. Notably, in vivo direct reprogramming of astrocytes into functional neurons were achieved in the adult mouse and non-human primate brains. In this review, we briefly summarized in vivo direct reprogramming of astrocytes into functional neurons as regenerative strategies for CNS diseases, mainly focusing on neurodegenerative diseases such as Parkinson’s disease (PD), Alzheimer’s disease (AD), and Huntington’s disease (HD). We highlight and outline the advantages and challenges of direct neuronal reprogramming from astrocytes in vivo for future neuroregenerative medicine.
Collapse
Affiliation(s)
- Youcui Wang
- Institute of Brain Science and Disease, School of Basic Medicine, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China
| | - Xiaoqin Zhang
- Institute of Brain Science and Disease, School of Basic Medicine, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China
| | - Fenghua Chen
- Institute of Brain Science and Disease, School of Basic Medicine, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China
| | - Ning Song
- Institute of Brain Science and Disease, School of Basic Medicine, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China
| | - Junxia Xie
- Institute of Brain Science and Disease, School of Basic Medicine, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China
| |
Collapse
|
15
|
Goldsteen PA, Yoseif C, Dolga AM, Gosens R. Human pluripotent stem cells for the modelling and treatment of respiratory diseases. Eur Respir Rev 2021; 30:30/161/210042. [PMID: 34348980 PMCID: PMC9488746 DOI: 10.1183/16000617.0042-2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 05/26/2021] [Indexed: 01/17/2023] Open
Abstract
Respiratory diseases are among the leading causes of morbidity and mortality worldwide, representing a major unmet medical need. New chemical entities rarely make it into the clinic to treat respiratory diseases, which is partially due to a lack of adequate predictive disease models and the limited availability of human lung tissues to model respiratory disease. Human pluripotent stem cells (hPSCs) may help fill this gap by serving as a scalable human in vitro model. In addition, human in vitro models of rare genetic mutations can be generated using hPSCs. hPSC-derived epithelial cells and organoids have already shown great potential for the understanding of disease mechanisms, for finding new potential targets by using high-throughput screening platforms, and for personalised treatments. These potentials can also be applied to other hPSC-derived lung cell types in the future. In this review, we will discuss how hPSCs have brought, and may continue to bring, major changes to the field of respiratory diseases by understanding the molecular mechanisms of the pathology and by finding efficient therapeutics. Human pluripotent stem cells may help to develop animal-free, fully human in vitro models to advance our understanding of disease mechanisms, for finding new potential targets by using high-throughput screening platforms, and for personalised treatments.https://bit.ly/3cahaqz
Collapse
Affiliation(s)
- Pien A Goldsteen
- Dept of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands .,GRIAC Research Institute, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Christina Yoseif
- Dept of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands
| | - Amalia M Dolga
- Dept of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands.,GRIAC Research Institute, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Reinoud Gosens
- Dept of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands.,GRIAC Research Institute, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
16
|
Zaki MM, Lesha E, Said K, Kiaee K, Robinson-McCarthy L, George H, Hanna A, Appleton E, Liu S, Ng AHM, Khoshakhlagh P, Church GM. Cell therapy strategies for COVID-19: Current approaches and potential applications. SCIENCE ADVANCES 2021; 7:eabg5995. [PMID: 34380619 PMCID: PMC8357240 DOI: 10.1126/sciadv.abg5995] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 06/22/2021] [Indexed: 05/03/2023]
Abstract
Coronavirus disease 2019 (COVID-19) continues to burden society worldwide. Despite most patients having a mild course, severe presentations have limited treatment options. COVID-19 manifestations extend beyond the lungs and may affect the cardiovascular, nervous, and other organ systems. Current treatments are nonspecific and do not address potential long-term consequences such as pulmonary fibrosis, demyelination, and ischemic organ damage. Cell therapies offer great potential in treating severe COVID-19 presentations due to their customizability and regenerative function. This review summarizes COVID-19 pathogenesis, respective areas where cell therapies have potential, and the ongoing 89 cell therapy trials in COVID-19 as of 1 January 2021.
Collapse
Affiliation(s)
- Mark M Zaki
- GC Therapeutics Inc., Cambridge, MA 02139, USA
- Department of Neurosurgery, University of Michigan, 1500 E Medical Center Dr., Ann Arbor, MI 48109, USA
| | - Emal Lesha
- GC Therapeutics Inc., Cambridge, MA 02139, USA
- Department of Neurosurgery, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Khaled Said
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Kiavash Kiaee
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | | | | | - Angy Hanna
- Department of Medicine, Beaumont Hospital, Royal Oak, MI, USA
| | - Evan Appleton
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02115, USA
| | - Songlei Liu
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02115, USA
| | - Alex H M Ng
- GC Therapeutics Inc., Cambridge, MA 02139, USA.
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02115, USA
| | - Parastoo Khoshakhlagh
- GC Therapeutics Inc., Cambridge, MA 02139, USA.
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02115, USA
| | - George M Church
- GC Therapeutics Inc., Cambridge, MA 02139, USA.
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02115, USA
| |
Collapse
|
17
|
Polyak A, Bankstahl JP, Besecke KFW, Hozsa C, Triebert W, Pannem RR, Manstein F, Borcholte T, Furch M, Zweigerdt R, Gieseler RK, Bengel FM, Ross TL. Simplified 89Zr-Labeling Protocol of Oxine (8-Hydroxyquinoline) Enabling Prolonged Tracking of Liposome-Based Nanomedicines and Cells. Pharmaceutics 2021; 13:1097. [PMID: 34371788 PMCID: PMC8309181 DOI: 10.3390/pharmaceutics13071097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/07/2021] [Accepted: 07/14/2021] [Indexed: 01/11/2023] Open
Abstract
In this work, a method for the preparation of the highly lipophilic labeling synthon [89Zr]Zr(oxinate)4 was optimized for the radiolabeling of liposomes and human induced pluripotent stem cells (hiPSCs). The aim was to establish a robust and reliable labeling protocol for enabling up to one week positron emission tomography (PET) tracing of lipid-based nanomedicines and transplanted or injected cells, respectively. [89Zr]Zr(oxinate)4 was prepared from oxine (8-hydroxyquinoline) and [89Zr]Zr(OH)2(C2O4). Earlier introduced liquid-liquid extraction methods were simplified by the optimization of buffering, pH, temperature and reaction times. For quality control, thin-layer chromatography (TLC), size-exclusion chromatography (SEC) and centrifugation were employed. Subsequently, the 89Zr-complex was incorporated into liposome formulations. PET/CT imaging of 89Zr-labeled liposomes was performed in healthy mice. Cell labeling was accomplished in PBS using suspensions of 3 × 106 hiPSCs, each. [89Zr]Zr(oxinate)4 was synthesized in very high radiochemical yields of 98.7% (96.8% ± 2.8%). Similarly, high internalization rates (≥90%) of [89Zr]Zr(oxinate)4 into liposomes were obtained over an 18 h incubation period. MicroPET and biodistribution studies confirmed the labeled nanocarriers' in vivo stability. Human iPSCs incorporated the labeling agent within 30 min with ~50% efficiency. Prolonged PET imaging is an ideal tool in the development of lipid-based nanocarriers for drug delivery and cell therapies. To this end, a reliable and reproducible 89Zr radiolabeling method was developed and tested successfully in a model liposome system and in hiPSCs alike.
Collapse
Affiliation(s)
- Andras Polyak
- Department of Nuclear Medicine, Hannover Medical School, 30625 Hannover, Germany; (J.P.B.); (F.M.B.); (T.L.R.)
| | - Jens P. Bankstahl
- Department of Nuclear Medicine, Hannover Medical School, 30625 Hannover, Germany; (J.P.B.); (F.M.B.); (T.L.R.)
| | - Karen F. W. Besecke
- Rodos Biotarget GmbH, Medical Park Hannover, 30625 Hannover, Germany; (K.F.W.B.); (C.H.); (R.R.P.); (T.B.); (M.F.); (R.K.G.)
- SolMic BioTech GmbH, 40225 Düsseldorf, Germany
| | - Constantin Hozsa
- Rodos Biotarget GmbH, Medical Park Hannover, 30625 Hannover, Germany; (K.F.W.B.); (C.H.); (R.R.P.); (T.B.); (M.F.); (R.K.G.)
| | - Wiebke Triebert
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), 30625 Hannover, Germany; (W.T.); (F.M.); (R.Z.)
- Department of Cardiac, Thoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical School, 30625 Hannover, Germany
| | - Rajeswara Rao Pannem
- Rodos Biotarget GmbH, Medical Park Hannover, 30625 Hannover, Germany; (K.F.W.B.); (C.H.); (R.R.P.); (T.B.); (M.F.); (R.K.G.)
- Bioloving GmbH & Co KG, 69126 Heidelberg, Germany
| | - Felix Manstein
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), 30625 Hannover, Germany; (W.T.); (F.M.); (R.Z.)
- Department of Cardiac, Thoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical School, 30625 Hannover, Germany
| | - Thomas Borcholte
- Rodos Biotarget GmbH, Medical Park Hannover, 30625 Hannover, Germany; (K.F.W.B.); (C.H.); (R.R.P.); (T.B.); (M.F.); (R.K.G.)
| | - Marcus Furch
- Rodos Biotarget GmbH, Medical Park Hannover, 30625 Hannover, Germany; (K.F.W.B.); (C.H.); (R.R.P.); (T.B.); (M.F.); (R.K.G.)
- SolMic BioTech GmbH, 40225 Düsseldorf, Germany
- Bioloving GmbH & Co KG, 69126 Heidelberg, Germany
| | - Robert Zweigerdt
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), 30625 Hannover, Germany; (W.T.); (F.M.); (R.Z.)
- Department of Cardiac, Thoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical School, 30625 Hannover, Germany
| | - Robert K. Gieseler
- Rodos Biotarget GmbH, Medical Park Hannover, 30625 Hannover, Germany; (K.F.W.B.); (C.H.); (R.R.P.); (T.B.); (M.F.); (R.K.G.)
- Department of Internal Medicine, and Laboratory of Immunology & Molecular Biology, University Hospital, Knappschaftskrankenhaus, Ruhr University Bochum, 44801 Bochum, Germany
| | - Frank M. Bengel
- Department of Nuclear Medicine, Hannover Medical School, 30625 Hannover, Germany; (J.P.B.); (F.M.B.); (T.L.R.)
| | - Tobias L. Ross
- Department of Nuclear Medicine, Hannover Medical School, 30625 Hannover, Germany; (J.P.B.); (F.M.B.); (T.L.R.)
| |
Collapse
|
18
|
Abstract
According to the recent patent filing trends, it has been observed that certain pharmaceutical technologies are more popular than others and are commonly referred to as emerging technologies. The emerging technologies in the pharmaceutical sector include artificial intelligence, big data and certain advanced biological therapies such as personalized medicine and stem cell therapy. These trends have various applications in the medicine and healthcare industry. Since these technologies are relatively new and each of them is very unique in its own way, current patent laws are inadequate to deal with the complex challenges associated with them. A brief analysis of the challenges associated with these emerging technologies and their applications is discussed in this paper.
Collapse
|
19
|
Kumar D, Talluri TR, Selokar NL, Hyder I, Kues WA. Perspectives of pluripotent stem cells in livestock. World J Stem Cells 2021; 13:1-29. [PMID: 33584977 PMCID: PMC7859985 DOI: 10.4252/wjsc.v13.i1.1] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/28/2020] [Accepted: 11/09/2020] [Indexed: 02/06/2023] Open
Abstract
The recent progress in derivation of pluripotent stem cells (PSCs) from farm animals opens new approaches not only for reproduction, genetic engineering, treatment and conservation of these species, but also for screening novel drugs for their efficacy and toxicity, and modelling of human diseases. Initial attempts to derive PSCs from the inner cell mass of blastocyst stages in farm animals were largely unsuccessful as either the cells survived for only a few passages, or lost their cellular potency; indicating that the protocols which allowed the derivation of murine or human embryonic stem (ES) cells were not sufficient to support the maintenance of ES cells from farm animals. This scenario changed by the innovation of induced pluripotency and by the development of the 3 inhibitor culture conditions to support naïve pluripotency in ES cells from livestock species. However, the long-term culture of livestock PSCs while maintaining the full pluripotency is still challenging, and requires further refinements. Here, we review the current achievements in the derivation of PSCs from farm animals, and discuss the potential application areas.
Collapse
Affiliation(s)
- Dharmendra Kumar
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Buffaloes, Hisar 125001, India.
| | - Thirumala R Talluri
- Equine Production Campus, ICAR-National Research Centre on Equines, Bikaner 334001, India
| | - Naresh L Selokar
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Buffaloes, Hisar 125001, India
| | - Iqbal Hyder
- Department of Physiology, NTR College of Veterinary Science, Gannavaram 521102, India
| | - Wilfried A Kues
- Department of Biotechnology, Friedrich-Loeffler-Institute, Federal Institute of Animal Health, Neustadt 31535, Germany
| |
Collapse
|
20
|
Yasui R, Sekine K, Yamaguchi K, Furukawa Y, Taniguchi H. Robust parameter design of human induced pluripotent stem cell differentiation protocols defines lineage-specific induction of anterior-posterior gut tube endodermal cells. Stem Cells 2021; 39:429-442. [PMID: 33400835 DOI: 10.1002/stem.3326] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 12/07/2020] [Indexed: 12/29/2022]
Abstract
Tissues and cells derived from pluripotent stem cells (PSC) are likely to become widely used in disease modeling, drug screening, and regenerative medicine. For these applications, the in vitro PSC differentiation process must be elaborately investigated and controlled to reliably obtain the desired end products. However, because traditional experimental methods, such as one factor at a time or brute-force approaches, are impractical for detailed screening of complex PSC cultivation conditions, more strategic and effective screening based on statistical design of experiments (DOE) ought to be indispensable. Among various DOE approaches, we regard robust parameter design (RPD) as particularly suited for differentiation protocol optimization due to its suitability for multifactorial screening. We confirmed the adaptability of RPD for investigating human induced PSC lineage specification toward anterior-posterior gut tube endodermal cells and clarified both the contribution of each cell signaling pathway and the effect of cell signaling condition alteration on marker RNA expression levels, while increasing the efficiency of the screening in 243-fold (18 vs 4374) compared with that of a brute-force approach. Specific induction of anterior foregut, hepatic, pancreatic, or mid-hindgut cells was achieved using seven iPSC strains with the optimal culture protocols established on the basis of RPD analysis. RPD has the potential to enable efficient construction and optimization of PSC differentiation protocols, and its use is recommended from fundamental research to mass production of PSC-derived products.
Collapse
Affiliation(s)
- Ryota Yasui
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan.,Fundamental Research Laboratory, Eiken Chemical Co., Ltd., Nogi, Tochigi, Japan
| | - Keisuke Sekine
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan.,Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Laboratory of Cancer Cell Systems, National Cancer Center Research Institute, Tokyo, Japan
| | - Kiyoshi Yamaguchi
- Division of Clinical Genome Research, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yoichi Furukawa
- Division of Clinical Genome Research, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hideki Taniguchi
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan.,Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Advanced Medical Research Center, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| |
Collapse
|
21
|
Renz PF, Spies D, Tsikrika P, Wutz A, Beyer TA, Ciaudo C. Inhibition of FGF and TGF-β Pathways in hESCs Identify STOX2 as a Novel SMAD2/4 Cofactor. BIOLOGY 2020; 9:biology9120470. [PMID: 33339109 PMCID: PMC7765495 DOI: 10.3390/biology9120470] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 12/15/2020] [Indexed: 01/10/2023]
Abstract
Simple Summary Signaling pathways are the means by which cells and tissue communicate, orchestrating key events during mammalian development, homeostasis, and disease. During development, signaling determines the identity of cells, and thereby controls morphogenesis and organ specification. Depending on the cellular context, these pathways can exert a broad range of even opposing functions. This is achieved, among other mechanisms, by crosstalk between pathways. Here, we examined how two pathways (the transforming growth factor-β (TGF-β) and the fibroblast growth factor (FGF)) cooperate in the maintenance and cell fate specification of human embryonic stem cells. We used inhibitory molecules for individual pathways on a short time series and analyzed the resulting variation in gene expression. In contrast to our expectations, we did not observe an extended crosstalk between the pathway at the gene regulatory level. However, we discovered STOX2 as a new primary target of the TGF-β signaling pathway. Our results show that STOX2 might act as a novel TGF-β signaling co-factor. Our work will contribute to understand how signaling by the TGF-β is mediated. In the future, these results might help to deepen our understanding of how signaling is propagated. Abstract The fibroblast growth factor (FGF) and the transforming growth factor-β (TGF-β) pathways are both involved in the maintenance of human embryonic stem cells (hESCs) and regulate the onset of their differentiation. Their converging functions have suggested that these pathways might share a wide range of overlapping targets. Published studies have focused on the long-term effects (24–48 h) of FGF and TGF-β inhibition in hESCs, identifying direct and indirect target genes. In this study, we focused on the earliest transcriptome changes occurring between 3 and 9 h after FGF and TGF-β inhibition to identify direct target genes only. Our analysis clearly shows that only a handful of target transcripts are common to both pathways. This is surprising in light of the previous literature, and has implications for models of cell signaling in human pluripotent cells. In addition, we identified STOX2 as a novel primary target of the TGF-β signaling pathway. We show that STOX2 might act as a novel SMAD2/4 cofactor. Taken together, our results provide insights into the effect of cell signaling on the transcription profile of human pluripotent cells
Collapse
Affiliation(s)
- Peter F. Renz
- Department of Biology, Swiss Federal Institute of Technology Zurich, Institute of Molecular Health Sciences, Otto-Stern Weg 7, CH-8093 Zurich, Switzerland; (P.F.R.); (D.S.); (P.T.); (A.W.)
- Molecular Life Science Program, Life Science Zurich Graduate School, Institute of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Daniel Spies
- Department of Biology, Swiss Federal Institute of Technology Zurich, Institute of Molecular Health Sciences, Otto-Stern Weg 7, CH-8093 Zurich, Switzerland; (P.F.R.); (D.S.); (P.T.); (A.W.)
- Molecular Life Science Program, Life Science Zurich Graduate School, Institute of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Panagiota Tsikrika
- Department of Biology, Swiss Federal Institute of Technology Zurich, Institute of Molecular Health Sciences, Otto-Stern Weg 7, CH-8093 Zurich, Switzerland; (P.F.R.); (D.S.); (P.T.); (A.W.)
- Molecular Life Science Program, Life Science Zurich Graduate School, Institute of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Anton Wutz
- Department of Biology, Swiss Federal Institute of Technology Zurich, Institute of Molecular Health Sciences, Otto-Stern Weg 7, CH-8093 Zurich, Switzerland; (P.F.R.); (D.S.); (P.T.); (A.W.)
| | - Tobias A. Beyer
- Department of Biology, Swiss Federal Institute of Technology Zurich, Institute of Molecular Health Sciences, Otto-Stern Weg 7, CH-8093 Zurich, Switzerland; (P.F.R.); (D.S.); (P.T.); (A.W.)
- Correspondence: (T.A.B.); (C.C.); Tel.: +41-44-633-08-58 (C.C.)
| | - Constance Ciaudo
- Department of Biology, Swiss Federal Institute of Technology Zurich, Institute of Molecular Health Sciences, Otto-Stern Weg 7, CH-8093 Zurich, Switzerland; (P.F.R.); (D.S.); (P.T.); (A.W.)
- Correspondence: (T.A.B.); (C.C.); Tel.: +41-44-633-08-58 (C.C.)
| |
Collapse
|
22
|
Shah S, Otsuka T, Bhattacharjee M, Laurencin CT. Minimally Invasive Cellular Therapies for Osteoarthritis Treatment. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2020. [DOI: 10.1007/s40883-020-00184-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
23
|
Petkov S, Dressel R, Rodriguez-Polo I, Behr R. Controlling the Switch from Neurogenesis to Pluripotency during Marmoset Monkey Somatic Cell Reprogramming with Self-Replicating mRNAs and Small Molecules. Cells 2020; 9:cells9112422. [PMID: 33167468 PMCID: PMC7694496 DOI: 10.3390/cells9112422] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 11/03/2020] [Indexed: 12/12/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) hold enormous potential for the development of cell-based therapies; however, the safety and efficacy of potential iPSC-based treatments need to be verified in relevant animal disease models before their application in the clinic. Here, we report the derivation of iPSCs from common marmoset monkeys (Callithrix jacchus) using self-replicating mRNA vectors based on the Venezuelan equine encephalitis virus (VEE-mRNAs). By transfection of marmoset fibroblasts with VEE-mRNAs carrying the human OCT4, KLF4, SOX2, and c-MYC and culture in the presence of small molecule inhibitors CHIR99021 and SB431542, we first established intermediate primary colonies with neural progenitor-like properties. In the second reprogramming step, we converted these colonies into transgene-free pluripotent stem cells by further culturing them with customized marmoset iPSC medium in feeder-free conditions. Our experiments revealed a novel paradigm for flexible reprogramming of somatic cells, where primary colonies obtained by a single VEE-mRNA transfection can be directed either toward the neural lineage or further reprogrammed to pluripotency. These results (1) will further enhance the role of the common marmoset as animal disease model for preclinical testing of iPSC-based therapies and (2) establish an in vitro system to experimentally address developmental signal transduction pathways in primates.
Collapse
Affiliation(s)
- Stoyan Petkov
- Platform Degenerative Diseases, German Primate Center, GmbH, Leibniz Institute for Primate Research, 37077 Göttingen, Germany;
- German Center for Cardiovascular Research (DZHK), partner site Göttingen, 37077 Göttingen, Germany;
- Correspondence: (S.P.); (R.B.); Tel.: +49-(0)551-3851-322 (S.P.); Tel.:+49-(0)551-3851-132 (R.B.)
| | - Ralf Dressel
- German Center for Cardiovascular Research (DZHK), partner site Göttingen, 37077 Göttingen, Germany;
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, 37077 Göttingen, Germany
| | - Ignacio Rodriguez-Polo
- Platform Degenerative Diseases, German Primate Center, GmbH, Leibniz Institute for Primate Research, 37077 Göttingen, Germany;
- German Center for Cardiovascular Research (DZHK), partner site Göttingen, 37077 Göttingen, Germany;
| | - Rüdiger Behr
- Platform Degenerative Diseases, German Primate Center, GmbH, Leibniz Institute for Primate Research, 37077 Göttingen, Germany;
- German Center for Cardiovascular Research (DZHK), partner site Göttingen, 37077 Göttingen, Germany;
- Correspondence: (S.P.); (R.B.); Tel.: +49-(0)551-3851-322 (S.P.); Tel.:+49-(0)551-3851-132 (R.B.)
| |
Collapse
|
24
|
Duckert B, Vinkx S, Braeken D, Fauvart M. Single-cell transfection technologies for cell therapies and gene editing. J Control Release 2020; 330:963-975. [PMID: 33160005 DOI: 10.1016/j.jconrel.2020.10.068] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 10/30/2020] [Accepted: 10/31/2020] [Indexed: 12/29/2022]
Abstract
Advances in gene editing and cell therapies have recently led to outstanding clinical successes. However, the lack of a cost-effective manufacturing process prevents the democratization of these innovative medical tools. Due to the common use of viral vectors, the step of transfection in which cells are engineered to gain new functions, is a major bottleneck in making safe and affordable cell products. A promising opportunity lies in Single-Cell Transfection Technologies (SCTTs). SCTTs have demonstrated higher efficiency, safety and scalability than conventional transfection methods. They can also feature unique abilities such as substantial dosage control over the cargo delivery, single-cell addressability and integration in microdevices comprising multiple monitoring modalities. Unfortunately, the potential of SCTTs is not fully appreciated: they are most often restricted to research settings with little adoption in clinical settings. To encourage their adoption, we review and compare recent developments in SCTTs, and how they can enable selected clinical applications. To help bridge the gap between fundamental research and its translation to the clinic, we also describe how Good Manufacturing Practices (GMP) can be integrated in the design of SCTTs.
Collapse
Affiliation(s)
- Bastien Duckert
- Department of Physics and Astronomy, KU Leuven, Celestijnenlaan 200d, 3001 Leuven, Belgium; IMEC, Kapeldreef 75, 3001 Leuven, Belgium.
| | | | | | | |
Collapse
|
25
|
Umezawa A, Sato Y, Kusakawa S, Amagase R, Akutsu H, Nakamura K, Kasahara M, Matsubara Y, Igarashi T. Research and Development Strategy for Future Embryonic Stem Cell-Based Therapy in Japan. JMA J 2020; 3:287-294. [PMID: 33225099 PMCID: PMC7676987 DOI: 10.31662/jmaj.2018-0029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 05/29/2020] [Indexed: 11/09/2022] Open
Abstract
Herewith, we review an updated progress of regenerative medical products using human embryonic stem cells (ESCs) in Japan. Two groups from Kyoto University and the National Center for Child Health and Development (NCCHD) established a novel derivation/cultivation system of ESCs for potential application in translational and clinical research. At the first stage of ESC derivation, murine feeder cells have been used in line with Japanese guidelines on public health associated with the implementation of the xenograft. To avoid exposure of ESCs to animal products in culture media, a xeno-free cultivating system has been established. Twelve ESCs (KhES-1, KhES-2, KhES-3, KhES-4, KhES-5, SEES-1, SEES-2, SEES-3, SEES-4, SEES-5, SEES-6, and SEES-7) are now available under a clinically relevant platform for industrially and clinically applicable regenerative medical products. NCCHD submitted an investigative new drug application to the Pharmaceuticals and Medical Devices Agency (PMDA) for using ESC-based products in patients with hyperammonemia due to genetic defects on March 2018 under the Pharmaceutical Affairs Law (now revised to the Pharmaceuticals, Medical Devices, and Other Therapeutic Products Act). Currently, up to ten ESC-based products are being prepared for intractable and rare disorders in Japan.
Collapse
Affiliation(s)
- Akihiro Umezawa
- Center for Regenerative Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Yoji Sato
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kawasaki, Japan
| | - Shinji Kusakawa
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kawasaki, Japan
| | - Rin Amagase
- Center for Regenerative Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Hidenori Akutsu
- Center for Regenerative Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Kazuaki Nakamura
- Center for Regenerative Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Mureo Kasahara
- Organ Transplantation Center, National Center for Child Health and Development, Tokyo, Japan
| | - Yoichi Matsubara
- National Center for Child Health and Development Research Institute, Tokyo, Japan
| | | |
Collapse
|
26
|
Somayaji R, Nichols DP, Bell SC. Cystic fibrosis - Ten promising therapeutic approaches in the current era of care. Expert Opin Investig Drugs 2020; 29:1107-1124. [PMID: 32744089 DOI: 10.1080/13543784.2020.1805733] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Cystic fibrosis (CF) is a genetic disease affecting multiple organ systems. Research and innovations in novel therapeutic agents and health care delivery have resulted in dramatic improvements in quality of life and survival for people with CF. Despite this, significant disease burden persists for many and this is compounded by disparities in treatment access and care which globally necessitates further work to improve outcomes. Because of the advent of numerous therapies which include gene-targeted modulators in parallel with specialized care delivery models, innovative efforts continue. AREAS COVERED In this review, we discuss the available data on investigational agents in clinical development and currently available treatments for CF. We also evaluate approaches to care delivery, consider treatment gaps, and propose future directions for advancement. EXPERT OPINION Since the discovery of the CF gene, CFTR modulators have provided a hallmark of success, even though it was thought not previously possible. This has led to reinvigorated efforts and innovations in treatment approaches and care delivery. Numerous challenges remain because of genetic and phenotypic heterogeneity, access issues, and therapeutic costs, but the collaborative approach between stakeholders for continued innovation fuels optimism. Abbreviations: CF cystic fibrosis; CFF Cystic Fibrosis Foundation (USA); CFTR cystic fibrosis transmembrane regulator; CRISPR clustered regularly interspaced short palindromic repeats; COX cyclo oxygenase; FDA US Food and Drug Administration; FEV1% forced expiratory volume in one second % predicted; F508del deletion of phenylalanine (F) in the 508th position (most common mutation); G551D substitution of the amino acid glycine by aspartate at position 551 in the nucleotide binding domain-1 of the CFTR gene; LMIC low- and middle-income country; LTB4 leukotriene B4; MDT multi-disciplinary care team; NO nitric oxide; NSAIDs non-steroidal anti-inflammatory drugs; SLPI secretory leukocyte protease inhibitor.
Collapse
Affiliation(s)
- Ranjani Somayaji
- Departments of Medicine; Microbiology, Immunology & Infectious Disease; Community Health Sciences, University of Calgary , Calgary, AB, Canada.,Snyder Institute for Chronic Diseases , Calgary, AB, Canada.,O'Brien Institute for Public Health , Calgary, AB, Canada
| | - Dave P Nichols
- Department of Pediatrics, Seattle Children's Hospital , Seattle, WA, USA.,Department of Pediatrics, University of Washington , Seattle, WA, USA.,Seattle Children's Research Institute , Seattle, WA, USA
| | - Scott C Bell
- Department of Thoracic Medicine, The Prince Charles Hospital , Brisbane, QLD, Australia.,Children's Health Research Centre, Faculty of Medicine, The University of Queensland , Brisbane, QLD, Australia.,Translational Research Institute , Brisbane, QLD, Australia
| |
Collapse
|
27
|
Protze SI, Lee JH, Keller GM. Human Pluripotent Stem Cell-Derived Cardiovascular Cells: From Developmental Biology to Therapeutic Applications. Cell Stem Cell 2020; 25:311-327. [PMID: 31491395 DOI: 10.1016/j.stem.2019.07.010] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Advances in our understanding of cardiovascular development have provided a roadmap for the directed differentiation of human pluripotent stem cells (hPSCs) to the major cell types found in the heart. In this Perspective, we review the state of the field in generating and maturing cardiovascular cells from hPSCs based on our fundamental understanding of heart development. We then highlight their applications for studying human heart development, modeling disease-performing drug screening, and cell replacement therapy. With the advancements highlighted here, the promise that hPSCs will deliver new treatments for degenerative and debilitating diseases may soon be fulfilled.
Collapse
Affiliation(s)
- Stephanie I Protze
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada.
| | - Jee Hoon Lee
- BlueRock Therapeutics ULC, Toronto, ON M5G 1L7, Canada
| | - Gordon M Keller
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada; Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 1L7, Canada.
| |
Collapse
|
28
|
Luciani M, Gritti A, Meneghini V. Human iPSC-Based Models for the Development of Therapeutics Targeting Neurodegenerative Lysosomal Storage Diseases. Front Mol Biosci 2020; 7:224. [PMID: 33062642 PMCID: PMC7530250 DOI: 10.3389/fmolb.2020.00224] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 08/10/2020] [Indexed: 01/30/2023] Open
Abstract
Lysosomal storage diseases (LSDs) are a group of rare genetic conditions. The absence or deficiency of lysosomal proteins leads to excessive storage of undigested materials and drives secondary pathological mechanisms including autophagy, calcium homeostasis, ER stress, and mitochondrial abnormalities. A large number of LSDs display mild to severe central nervous system (CNS) involvement. Animal disease models and post-mortem tissues partially recapitulate the disease or represent the final stage of CNS pathology, respectively. In the last decades, human models based on induced pluripotent stem cells (hiPSCs) have been extensively applied to investigate LSD pathology in several tissues and organs, including the CNS. Neural stem/progenitor cells (NSCs) derived from patient-specific hiPSCs (hiPS-NSCs) are a promising tool to define the effects of the pathological storage on neurodevelopment, survival and function of neurons and glial cells in neurodegenerative LSDs. Additionally, the development of novel 2D co-culture systems and 3D hiPSC-based models is fostering the investigation of neuron-glia functional and dysfunctional interactions, also contributing to define the role of neurodevelopment and neuroinflammation in the onset and progression of the disease, with important implications in terms of timing and efficacy of treatments. Here, we discuss the advantages and limits of the application of hiPS-NSC-based models in the study and treatment of CNS pathology in different LSDs. Additionally, we review the state-of-the-art and the prospective applications of NSC-based therapy, highlighting the potential exploitation of hiPS-NSCs for gene and cell therapy approaches in the treatment of neurodegenerative LSDs.
Collapse
Affiliation(s)
- Marco Luciani
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Angela Gritti
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Vasco Meneghini
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
29
|
Upadhya R, Madhu LN, Attaluri S, Gitaí DLG, Pinson MR, Kodali M, Shetty G, Zanirati G, Kumar S, Shuai B, Weintraub ST, Shetty AK. Extracellular vesicles from human iPSC-derived neural stem cells: miRNA and protein signatures, and anti-inflammatory and neurogenic properties. J Extracell Vesicles 2020; 9:1809064. [PMID: 32944193 PMCID: PMC7480597 DOI: 10.1080/20013078.2020.1809064] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Grafting of neural stem cells (NSCs) derived from human induced pluripotent stem cells (hiPSCs) has shown promise for brain repair after injury or disease, but safety issues have hindered their clinical application. Employing nano-sized extracellular vesicles (EVs) derived from hiPSC-NSCs appears to be a safer alternative because they likely have similar neuroreparative properties as NSCs and are amenable for non-invasive administration as an autologous or allogeneic off-the-shelf product. However, reliable methods for isolation, characterization and testing the biological properties of EVs are critically needed for translation. We investigated signatures of miRNAs and proteins and the biological activity of EVs, isolated from hiPSC-NSCs through a combination of anion-exchange chromatography (AEC) and size-exclusion chromatography (SEC). AEC and SEC facilitated the isolation of EVs with intact ultrastructure and expressing CD9, CD63, CD81, ALIX and TSG 101. Small RNA sequencing, proteomic analysis, pathway analysis and validation of select miRNAs and proteins revealed that EVs were enriched with miRNAs and proteins involved in neuroprotective, anti-apoptotic, antioxidant, anti-inflammatory, blood-brain barrier repairing, neurogenic and Aβ reducing activities. Besides, EVs comprised miRNAs and/or proteins capable of promoting synaptogenesis, synaptic plasticity and better cognitive function. Investigations using an in vitro macrophage assay and a mouse model of status epilepticus confirmed the anti-inflammatory activity of EVs. Furthermore, the intranasal administration of EVs resulted in the incorporation of EVs by neurons, microglia and astrocytes in virtually all adult rat and mouse brain regions, and enhancement of hippocampal neurogenesis. Thus, biologically active EVs containing miRNAs and proteins relevant to brain repair could be isolated from hiPSC-NSC cultures, making them a suitable biologic for treating neurodegenerative disorders.
Collapse
Affiliation(s)
- Raghavendra Upadhya
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, Texas, USA
| | - Leelavathi N Madhu
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, Texas, USA
| | - Sahithi Attaluri
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, Texas, USA
| | - Daniel Leite Góes Gitaí
- Department of Cellular and Molecular Biology, Institute of Biological Sciences and Health, Federal University of Alagoas, Brazil
| | - Marisa R Pinson
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, Texas, USA
| | - Maheedhar Kodali
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, Texas, USA
| | - Geetha Shetty
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, Texas, USA
| | - Gabriele Zanirati
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, Texas, USA
| | - Smrithi Kumar
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, Texas, USA
| | - Bing Shuai
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, Texas, USA
| | - Susan T Weintraub
- Department of Biochemistry and Structural Biology, UT Health San Antonio, San Antonio, Texas, USA
| | - Ashok K Shetty
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, Texas, USA
| |
Collapse
|
30
|
Characterization of Endoplasmic Reticulum (ER) in Human Pluripotent Stem Cells Revealed Increased Susceptibility to Cell Death upon ER Stress. Cells 2020; 9:cells9051078. [PMID: 32357563 PMCID: PMC7291192 DOI: 10.3390/cells9051078] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/22/2020] [Accepted: 04/23/2020] [Indexed: 12/19/2022] Open
Abstract
Human pluripotent stem cells (hPSCs), such as embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), have a well-orchestrated program for differentiation and self-renewal. However, the structural features of unique proteostatic-maintaining mechanisms in hPSCs and their features, distinct from those of differentiated cells, in response to cellular stress remain unclear. We evaluated and compared the morphological features and stress response of hPSCs and fibroblasts. Compared to fibroblasts, electron microscopy showed simpler/fewer structures with fewer networks in the endoplasmic reticulum (ER) of hPSCs, as well as lower expression of ER-related genes according to meta-analysis. As hPSCs contain low levels of binding immunoglobulin protein (BiP), an ER chaperone, thapsigargin treatment sharply increased the gene expression of the unfolded protein response. Thus, hPSCs with decreased chaperone function reacted sensitively to ER stress and entered apoptosis faster than fibroblasts. Such ER stress-induced apoptotic processes were abolished by tauroursodeoxycholic acid, an ER-stress reliever. Hence, our results revealed that as PSCs have an underdeveloped structure and express fewer BiP chaperone proteins than somatic cells, they are more susceptible to ER stress-induced apoptosis in response to stress.
Collapse
|
31
|
Lipus A, Janosz E, Ackermann M, Hetzel M, Dahlke J, Buchegger T, Wunderlich S, Martin U, Cathomen T, Schambach A, Moritz T, Lachmann N. Targeted Integration of Inducible Caspase-9 in Human iPSCs Allows Efficient in vitro Clearance of iPSCs and iPSC-Macrophages. Int J Mol Sci 2020; 21:E2481. [PMID: 32260086 PMCID: PMC7177583 DOI: 10.3390/ijms21072481] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 03/30/2020] [Accepted: 04/01/2020] [Indexed: 02/06/2023] Open
Abstract
Induced pluripotent stem cells (iPSCs) offer great promise for the field of regenerative medicine, and iPSC-derived cells have already been applied in clinical practice. However, potential contamination of effector cells with residual pluripotent cells (e.g., teratoma-initiating cells) or effector cell-associated side effects may limit this approach. This also holds true for iPSC-derived hematopoietic cells. Given the therapeutic benefit of macrophages in different disease entities and the feasibility to derive macrophages from human iPSCs, we established human iPSCs harboring the inducible Caspase-9 (iCasp9) suicide safety switch utilizing transcription activator-like effector nuclease (TALEN)-based designer nuclease technology. Mono- or bi-allelic integration of the iCasp9 gene cassette into the AAVS1 locus showed no effect on the pluripotency of human iPSCs and did not interfere with their differentiation towards macrophages. In both, iCasp9-mono and iCasp9-bi-allelic clones, concentrations of 0.1 nM AP20187 were sufficient to induce apoptosis in more than 98% of iPSCs and their progeny-macrophages. Thus, here we provide evidence that the introduction of the iCasp9 suicide gene into the AAVS1 locus enables the effective clearance of human iPSCs and thereof derived macrophages.
Collapse
Affiliation(s)
- Alexandra Lipus
- RG Reprogramming and Gene Therapy, Hannover Medical School, Hannover 30625, Germany; (A.L.); (E.J.); (M.H.); (T.M.)
- Institute of Experimental Hematology, REBIRTH, Hannover Medical School, Hannover 30625, Germany; (M.A.); (J.D.); (T.B.); (A.S.)
| | - Ewa Janosz
- RG Reprogramming and Gene Therapy, Hannover Medical School, Hannover 30625, Germany; (A.L.); (E.J.); (M.H.); (T.M.)
- Institute of Experimental Hematology, REBIRTH, Hannover Medical School, Hannover 30625, Germany; (M.A.); (J.D.); (T.B.); (A.S.)
| | - Mania Ackermann
- Institute of Experimental Hematology, REBIRTH, Hannover Medical School, Hannover 30625, Germany; (M.A.); (J.D.); (T.B.); (A.S.)
- RG Translational Hematology of Congenital Diseases, Hannover Medical School, Hannover 30625, Germany
| | - Miriam Hetzel
- RG Reprogramming and Gene Therapy, Hannover Medical School, Hannover 30625, Germany; (A.L.); (E.J.); (M.H.); (T.M.)
- Institute of Experimental Hematology, REBIRTH, Hannover Medical School, Hannover 30625, Germany; (M.A.); (J.D.); (T.B.); (A.S.)
| | - Julia Dahlke
- Institute of Experimental Hematology, REBIRTH, Hannover Medical School, Hannover 30625, Germany; (M.A.); (J.D.); (T.B.); (A.S.)
| | - Theresa Buchegger
- Institute of Experimental Hematology, REBIRTH, Hannover Medical School, Hannover 30625, Germany; (M.A.); (J.D.); (T.B.); (A.S.)
- RG Translational Hematology of Congenital Diseases, Hannover Medical School, Hannover 30625, Germany
| | - Stephanie Wunderlich
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, REBIRTH, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Hannover 30625, Germany; (S.W.); (U.M.)
| | - Ulrich Martin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, REBIRTH, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Hannover 30625, Germany; (S.W.); (U.M.)
| | - Toni Cathomen
- Institute for Transfusion Medicine and Gene Therapy, Medical Center-University of Freiburg, Freiburg 79106, Germany;
- Center for Chronic Immunodeficiency, Faculty of Medicine, University of Freiburg, Freiburg 79095, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, REBIRTH, Hannover Medical School, Hannover 30625, Germany; (M.A.); (J.D.); (T.B.); (A.S.)
| | - Thomas Moritz
- RG Reprogramming and Gene Therapy, Hannover Medical School, Hannover 30625, Germany; (A.L.); (E.J.); (M.H.); (T.M.)
- Institute of Experimental Hematology, REBIRTH, Hannover Medical School, Hannover 30625, Germany; (M.A.); (J.D.); (T.B.); (A.S.)
| | - Nico Lachmann
- Institute of Experimental Hematology, REBIRTH, Hannover Medical School, Hannover 30625, Germany; (M.A.); (J.D.); (T.B.); (A.S.)
- RG Translational Hematology of Congenital Diseases, Hannover Medical School, Hannover 30625, Germany
| |
Collapse
|
32
|
Collino F, Lopes JA, Tapparo M, Tortelote GG, Kasai-Brunswick TH, Lopes GM, Almeida DB, Skovronova R, Wendt CHC, de Miranda KR, Bussolati B, Vieyra A, Lindoso RS. Extracellular Vesicles Derived from Induced Pluripotent Stem Cells Promote Renoprotection in Acute Kidney Injury Model. Cells 2020; 9:cells9020453. [PMID: 32079274 PMCID: PMC7072760 DOI: 10.3390/cells9020453] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 01/16/2020] [Accepted: 02/12/2020] [Indexed: 12/22/2022] Open
Abstract
Induced pluripotent stem cells (iPSC) have been the focus of several studies due to their wide range of application, including in cellular therapy. The use of iPSC in regenerative medicine is limited by their tumorigenic potential. Extracellular vesicles (EV) derived from stem cells have been shown to support renal recovery after injury. However, no investigation has explored the potential of iPSC-EV in the treatment of kidney diseases. To evaluate this potential, we submitted renal tubule cells to hypoxia-reoxygenation injury, and we analyzed cell death rate and changes in functional mitochondria mass. An in vivo model of ischemia-reperfusion injury was used to evaluate morphological and functional alterations. Gene array profile was applied to investigate the mechanism involved in iPSC-EV effects. In addition, EV derived from adipose mesenchymal cells (ASC-EV) were also used to compare the potential of iPSC-EV in support of tissue recovery. The results showed that iPSC-EV were capable of reducing cell death and inflammatory response with similar efficacy than ASC-EV. Moreover, iPSC-EV protected functional mitochondria and regulated several genes associated with oxidative stress. Taken together, these results show that iPSC can be an alternative source of EV in the treatment of different aspects of kidney disease.
Collapse
Affiliation(s)
- Federica Collino
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil; (F.C.); (J.A.L.); (G.G.T.); (T.H.K.-B.); (G.M.C.L.); (D.B.A.)
- National Institute of Science and Technology for Regenerative Medicine-REGENERA, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
- Department of Biomedical Sciences, University of Padova, 35131 Padua, Italy
| | - Jarlene A. Lopes
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil; (F.C.); (J.A.L.); (G.G.T.); (T.H.K.-B.); (G.M.C.L.); (D.B.A.)
- National Institute of Science and Technology for Regenerative Medicine-REGENERA, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
- National Center for Structural Biology and Bioimaging/CENABIO, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Marta Tapparo
- Department of Medical Sciences, Molecular Biotechnology Center, University of Torino, 10126 Torino, Italy;
| | - Giovane G. Tortelote
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil; (F.C.); (J.A.L.); (G.G.T.); (T.H.K.-B.); (G.M.C.L.); (D.B.A.)
- Department of Pediatrics’ Section of Pediatric Nephrology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Taís H. Kasai-Brunswick
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil; (F.C.); (J.A.L.); (G.G.T.); (T.H.K.-B.); (G.M.C.L.); (D.B.A.)
- National Institute of Science and Technology for Regenerative Medicine-REGENERA, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
- National Center for Structural Biology and Bioimaging/CENABIO, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Gustavo M.C. Lopes
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil; (F.C.); (J.A.L.); (G.G.T.); (T.H.K.-B.); (G.M.C.L.); (D.B.A.)
- National Institute of Science and Technology for Regenerative Medicine-REGENERA, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
- National Center for Structural Biology and Bioimaging/CENABIO, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Douglas B. Almeida
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil; (F.C.); (J.A.L.); (G.G.T.); (T.H.K.-B.); (G.M.C.L.); (D.B.A.)
- National Institute of Science and Technology for Regenerative Medicine-REGENERA, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
- National Center for Structural Biology and Bioimaging/CENABIO, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Renata Skovronova
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Turin, Italy (B.B.)
| | - Camila H. C. Wendt
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil; (F.C.); (J.A.L.); (G.G.T.); (T.H.K.-B.); (G.M.C.L.); (D.B.A.)
| | - Kildare R. de Miranda
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil; (F.C.); (J.A.L.); (G.G.T.); (T.H.K.-B.); (G.M.C.L.); (D.B.A.)
- National Center for Structural Biology and Bioimaging/CENABIO, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
- National Institute of Science and Technology of Structural Biology and Bioimaging-INBEB, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Turin, Italy (B.B.)
| | - Adalberto Vieyra
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil; (F.C.); (J.A.L.); (G.G.T.); (T.H.K.-B.); (G.M.C.L.); (D.B.A.)
- National Institute of Science and Technology for Regenerative Medicine-REGENERA, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
- National Center for Structural Biology and Bioimaging/CENABIO, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
- Graduate Program of Translational Biomedicine/BIOTRANS, Grande Rio University, 25071-202 Duque de Caxias, Brazil
- Correspondence: (A.V.); (R.S.L.); Tel.: +55-21-3938-6521 (A.V.); +55-21-3938-6520 (R.S.L.)
| | - Rafael Soares Lindoso
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil; (F.C.); (J.A.L.); (G.G.T.); (T.H.K.-B.); (G.M.C.L.); (D.B.A.)
- National Institute of Science and Technology for Regenerative Medicine-REGENERA, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
- National Center for Structural Biology and Bioimaging/CENABIO, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
- Correspondence: (A.V.); (R.S.L.); Tel.: +55-21-3938-6521 (A.V.); +55-21-3938-6520 (R.S.L.)
| |
Collapse
|
33
|
Tambuyzer E, Vandendriessche B, Austin CP, Brooks PJ, Larsson K, Miller Needleman KI, Valentine J, Davies K, Groft SC, Preti R, Oprea TI, Prunotto M. Therapies for rare diseases: therapeutic modalities, progress and challenges ahead. Nat Rev Drug Discov 2019; 19:93-111. [PMID: 31836861 DOI: 10.1038/s41573-019-0049-9] [Citation(s) in RCA: 203] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2019] [Indexed: 12/26/2022]
Abstract
Most rare diseases still lack approved treatments despite major advances in research providing the tools to understand their molecular basis, as well as legislation providing regulatory and economic incentives to catalyse the development of specific therapies. Addressing this translational gap is a multifaceted challenge, for which a key aspect is the selection of the optimal therapeutic modality for translating advances in rare disease knowledge into potential medicines, known as orphan drugs. With this in mind, we discuss here the technological basis and rare disease applicability of the main therapeutic modalities, including small molecules, monoclonal antibodies, protein replacement therapies, oligonucleotides and gene and cell therapies, as well as drug repurposing. For each modality, we consider its strengths and limitations as a platform for rare disease therapy development and describe clinical progress so far in developing drugs based on it. We also discuss selected overarching topics in the development of therapies for rare diseases, such as approval statistics, engagement of patients in the process, regulatory pathways and digital tools.
Collapse
Affiliation(s)
- Erik Tambuyzer
- BioPontis Alliance for Rare Diseases Foundation fup/son, Brussels, Belgium. .,BioPontis Alliance Rare Disease Foundation, Inc, Raleigh, NC, USA.
| | - Benjamin Vandendriessche
- Byteflies, Antwerp, Belgium.,Department of Electrical, Computer, and Systems Engineering (ECSE), Case Western Reserve University, Cleveland, OH, USA
| | - Christopher P Austin
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Philip J Brooks
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Kristina Larsson
- Orphan Medicines Office, European Medicines Agency, Amsterdam, Netherlands
| | | | | | - Kay Davies
- MDUK Oxford Neuromuscular Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Stephen C Groft
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Robert Preti
- Hitachi Chemical Regenerative Medicine Business Sector, Allendale, NJ, USA
| | - Tudor I Oprea
- Translational Informatics Division, Department of Internal Medicine, University of New Mexico Albuquerque, Albuquerque, NM, USA.,UNM Comprehensive Cancer Center, University of New Mexico Health Science Center, Albuquerque, NM, USA
| | - Marco Prunotto
- School of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
34
|
Ortuño-Costela MDC, Cerrada V, García-López M, Gallardo ME. The Challenge of Bringing iPSCs to the Patient. Int J Mol Sci 2019; 20:E6305. [PMID: 31847153 PMCID: PMC6940848 DOI: 10.3390/ijms20246305] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 12/10/2019] [Accepted: 12/11/2019] [Indexed: 12/13/2022] Open
Abstract
The implementation of induced pluripotent stem cells (iPSCs) in biomedical research more than a decade ago, resulted in a huge leap forward in the highly promising area of personalized medicine. Nowadays, we are even closer to the patient than ever. To date, there are multiple examples of iPSCs applications in clinical trials and drug screening. However, there are still many obstacles to overcome. In this review, we will focus our attention on the advantages of implementing induced pluripotent stem cells technology into the clinics but also commenting on all the current drawbacks that could hinder this promising path towards the patient.
Collapse
Affiliation(s)
- María del Carmen Ortuño-Costela
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Madrid, Spain. Instituto de Investigaciones Biomédicas “Alberto Sols”, (UAM-CSIC), 28029 Madrid, Spain;
- Grupo de Investigación Traslacional con células iPS, Instituto de Investigación Sanitaria Hospital 12 de Octubre (i+12), 28041 Madrid, Spain; (V.C.); (M.G.-L.)
| | - Victoria Cerrada
- Grupo de Investigación Traslacional con células iPS, Instituto de Investigación Sanitaria Hospital 12 de Octubre (i+12), 28041 Madrid, Spain; (V.C.); (M.G.-L.)
| | - Marta García-López
- Grupo de Investigación Traslacional con células iPS, Instituto de Investigación Sanitaria Hospital 12 de Octubre (i+12), 28041 Madrid, Spain; (V.C.); (M.G.-L.)
| | - M. Esther Gallardo
- Grupo de Investigación Traslacional con células iPS, Instituto de Investigación Sanitaria Hospital 12 de Octubre (i+12), 28041 Madrid, Spain; (V.C.); (M.G.-L.)
- Centro de Investigación Biomédica en Red (CIBERER), 28029 Madrid, Spain
| |
Collapse
|
35
|
Chemically-Defined, Xeno-Free, Scalable Production of hPSC-Derived Definitive Endoderm Aggregates with Multi-Lineage Differentiation Potential. Cells 2019; 8:cells8121571. [PMID: 31817235 PMCID: PMC6953099 DOI: 10.3390/cells8121571] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 11/29/2019] [Accepted: 12/03/2019] [Indexed: 12/30/2022] Open
Abstract
For the production and bio-banking of differentiated derivatives from human pluripotent stem cells (hPSCs) in large quantities for drug screening and cellular therapies, well-defined and robust procedures for differentiation and cryopreservation are required. Definitive endoderm (DE) gives rise to respiratory and digestive epithelium, as well as thyroid, thymus, liver, and pancreas. Here, we present a scalable, universal process for the generation of DE from human-induced pluripotent stem cells (hiPSCs) and embryonic stem cells (hESCs). Optimal control during the differentiation process was attained in chemically-defined and xeno-free suspension culture, and high flexibility of the workflow was achieved by the introduction of an efficient cryopreservation step at the end of DE differentiation. DE aggregates were capable of differentiating into hepatic-like, pancreatic, intestinal, and lung progenitor cells. Scale-up of the differentiation process using stirred-tank bioreactors enabled production of large quantities of DE aggregates. This process provides a useful advance for versatile applications of DE lineages, in particular for cell therapies and drug screening.
Collapse
|
36
|
Bogomiakova ME, Eremeev AV, Lagarkova MA. At Home among Strangers: Is It Possible to Create Hypoimmunogenic Pluripotent Stem Cell Lines? Mol Biol 2019. [DOI: 10.1134/s0026893319050042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
37
|
Zavala G, Sandoval C, Meza D, Contreras R, Gubelin W, Khoury M. Differentiation of adipose-derived stem cells to functional CD105 neg CD73 low melanocyte precursors guided by defined culture condition. Stem Cell Res Ther 2019; 10:249. [PMID: 31399041 PMCID: PMC6688240 DOI: 10.1186/s13287-019-1364-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 07/26/2019] [Accepted: 07/29/2019] [Indexed: 12/18/2022] Open
Abstract
Background The generation of functional human epidermal melanocytes (HEM) from stem cells provides an unprecedented source for cell-based therapy in vitiligo. Despite the important efforts exerted to obtain melanin-producing cells from stem cells, pre-clinical results still lack the safety and scalability characteristics essential for their translational application. Methods Here, we report a rapid and efficient protocol based on defined culture conditions capable of differentiating adult adipose-derived stem cells (ADSC) to scalable amounts of proliferative melanocyte precursors (PreMel) within 30 days. PreMel were characterized in vitro through qPCR, Western blot, flow cytometry, biochemical assays, and in vivo assays in immunocompromised mice (NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ, or NSG). Results After 30 days of differentiation, the stem cell-derived PreMel were defined as CD105neg CD73low according to immunophenotypic changes in comparison with parental stem cell markers. In addition, expression of microphthalmia-associated transcription factor (MITF), active tyrosinase (TYR), and the terminal differentiation-involved premelanosome protein (PMEL) were detected. Furthermore, PreMel had the potential to synthesize melanin and package it into melanosomes both in vitro and in vivo in NSG mice skin. Conclusions This study proposes a rapid and scalable protocol for the generation of proliferative melanocyte precursors (PreMel) from ADSC. These PreMel display the essential functional characteristics of bona fide HEM, opening a new path for an autologous cellular therapy for vitiligo patients. Electronic supplementary material The online version of this article (10.1186/s13287-019-1364-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Gabriela Zavala
- Consorcio Regenero, La Plaza 2501, Las Condes, Santiago, Chile.,Biomedical Research Center (CIB), Faculty of Medicine, Universidad de los Andes, Monseñor Álvaro del Portillo 12455, Las Condes, Santiago, Chile
| | - Carolina Sandoval
- Biomedical Research Center (CIB), Faculty of Medicine, Universidad de los Andes, Monseñor Álvaro del Portillo 12455, Las Condes, Santiago, Chile
| | - Daniel Meza
- Consorcio Regenero, La Plaza 2501, Las Condes, Santiago, Chile.,Biomedical Research Center (CIB), Faculty of Medicine, Universidad de los Andes, Monseñor Álvaro del Portillo 12455, Las Condes, Santiago, Chile
| | - Rafael Contreras
- Cells for Cells, La Plaza 2501, Las Condes, Santiago, Chile.,Biomedical Research Center (CIB), Faculty of Medicine, Universidad de los Andes, Monseñor Álvaro del Portillo 12455, Las Condes, Santiago, Chile
| | - Walter Gubelin
- Faculty of Medicine, Universidad de los Andes, Monseñor Álvaro del Portillo 12455, Las Condes, Santiago, Chile
| | - Maroun Khoury
- Consorcio Regenero, La Plaza 2501, Las Condes, Santiago, Chile. .,Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de los Andes, Monseñor Álvaro del Portillo 12455, Las Condes, Santiago, Chile. .,Cells for Cells, La Plaza 2501, Las Condes, Santiago, Chile. .,Biomedical Research Center (CIB), Faculty of Medicine, Universidad de los Andes, Monseñor Álvaro del Portillo 12455, Las Condes, Santiago, Chile.
| |
Collapse
|
38
|
Alia C, Terrigno M, Busti I, Cremisi F, Caleo M. Pluripotent Stem Cells for Brain Repair: Protocols and Preclinical Applications in Cortical and Hippocampal Pathologies. Front Neurosci 2019; 13:684. [PMID: 31447623 PMCID: PMC6691396 DOI: 10.3389/fnins.2019.00684] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 06/14/2019] [Indexed: 12/13/2022] Open
Abstract
Brain injuries causing chronic sensory or motor deficit, such as stroke, are among the leading causes of disability worldwide, according to the World Health Organization; furthermore, they carry heavy social and economic burdens due to decreased quality of life and need of assistance. Given the limited effectiveness of rehabilitation, novel therapeutic strategies are required to enhance functional recovery. Since cell-based approaches have emerged as an intriguing and promising strategy to promote brain repair, many efforts have been made to study the functional integration of neurons derived from pluripotent stem cells (PSCs), or fetal neurons, after grafting into the damaged host tissue. PSCs hold great promises for their clinical applications, such as cellular replacement of damaged neural tissues with autologous neurons. They also offer the possibility to create in vitro models to assess the efficacy of drugs and therapies. Notwithstanding these potential applications, PSC-derived transplanted neurons have to match the precise sub-type, positional and functional identity of the lesioned neural tissue. Thus, the requirement of highly specific and efficient differentiation protocols of PSCs in neurons with appropriate neural identity constitutes the main challenge limiting the clinical use of stem cells in the near future. In this Review, we discuss the recent advances in the derivation of telencephalic (cortical and hippocampal) neurons from PSCs, assessing specificity and efficiency of the differentiation protocols, with particular emphasis on the genetic and molecular characterization of PSC-derived neurons. Second, we address the remaining challenges for cellular replacement therapies in cortical brain injuries, focusing on electrophysiological properties, functional integration and therapeutic effects of the transplanted neurons.
Collapse
Affiliation(s)
- Claudia Alia
- CNR Neuroscience Institute, National Research Council (CNR), Pisa, Italy
| | - Marco Terrigno
- Laboratory of Biology, Scuola Normale Superiore, Pisa, Italy
| | - Irene Busti
- CNR Neuroscience Institute, National Research Council (CNR), Pisa, Italy.,Department of Neuroscience, Psychology, Drugs and Child Health Area, School of Psychology, University of Florence, Florence, Italy
| | - Federico Cremisi
- Laboratory of Biology, Scuola Normale Superiore, Pisa, Italy.,Biophysics Institute (IBF), National Research Council (CNR), Pisa, Italy
| | - Matteo Caleo
- CNR Neuroscience Institute, National Research Council (CNR), Pisa, Italy.,Department of Biomedical Sciences, University of Padua, Padua, Italy.,Padua Neuroscience Center, University of Padua, Padua, Italy
| |
Collapse
|
39
|
El Wazan L, Urrutia-Cabrera D, Wong RCB. Using transcription factors for direct reprogramming of neurons in vitro. World J Stem Cells 2019; 11:431-444. [PMID: 31396370 PMCID: PMC6682505 DOI: 10.4252/wjsc.v11.i7.431] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 06/07/2019] [Accepted: 06/27/2019] [Indexed: 02/06/2023] Open
Abstract
Cell therapy offers great promises in replacing the neurons lost due to neurodegenerative diseases or injuries. However, a key challenge is the cellular source for transplantation which is often limited by donor availability. Direct reprogramming provides an exciting avenue to generate specialized neuron subtypes in vitro, which have the potential to be used for autologous transplantation, as well as generation of patient-specific disease models in the lab for drug discovery and testing gene therapy. Here we present a detailed review on transcription factors that promote direct reprogramming of specific neuronal subtypes with particular focus on glutamatergic, GABAergic, dopaminergic, sensory and retinal neurons. We will discuss the developmental role of master transcriptional regulators and specification factors for neuronal subtypes, and summarize their use in promoting direct reprogramming into different neuronal subtypes. Furthermore, we will discuss up-and-coming technologies that advance the cell reprogramming field, including the use of computational prediction of reprogramming factors, opportunity of cellular reprogramming using small chemicals and microRNA, as well as the exciting potential for applying direct reprogramming in vivo as a novel approach to promote neuro-regeneration within the body. Finally, we will highlight the clinical potential of direct reprogramming and discuss the hurdles that need to be overcome for clinical translation.
Collapse
Affiliation(s)
- Layal El Wazan
- Cellular Reprogramming Unit, Centre for Eye Research Australia, Melbourne 3004, Australia
| | - Daniel Urrutia-Cabrera
- Cellular Reprogramming Unit, Centre for Eye Research Australia, Melbourne 3004, Australia
| | | |
Collapse
|
40
|
Hunt CJ. Technical Considerations in the Freezing, Low-Temperature Storage and Thawing of Stem Cells for Cellular Therapies. Transfus Med Hemother 2019; 46:134-150. [PMID: 31244583 PMCID: PMC6558338 DOI: 10.1159/000497289] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 01/26/2019] [Indexed: 12/31/2022] Open
Abstract
The commercial and clinical development of cellular therapy products will invariably require cryopreservation and frozen storage of cellular starting materials, intermediates and/or final product. Optimising cryopreservation is as important as optimisation of the cell culture process in obtaining maximum yield and a consistent end-product. Suboptimal cryopreservation can lead not only to batch-to-batch variation, lowered cellular functionality and reduced cell yield, but also to the potential selection of subpopulations with genetic or epigenetic characteristics divergent from the original cell line. Regulatory requirements also impact on cryopreservation as these will require a robust and reproducible approach to the freezing, storage and thawing of the product. This requires attention to all aspects of the application of low temperatures: from the choice of freezing container and cryoprotectant, the cooling rate employed and its mode of de-livery, the correct handling of the frozen material during storage and transportation, to the eventual thawing of the product by the end-user. Each of these influences all of the others to a greater or lesser extent and none should be ignored. This paper seeks to provide practical insights and alternative solutions to the technical challenges faced during cryopreservation of cells for use in cellular therapies.
Collapse
|
41
|
Thesleff I. From understanding tooth development to bioengineering of teeth. Eur J Oral Sci 2019; 126 Suppl 1:67-71. [PMID: 30178557 DOI: 10.1111/eos.12421] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2018] [Indexed: 12/30/2022]
Abstract
Remarkable breakthroughs in the fields of developmental biology and stem cell biology during the last 15 yr have led to a new level of understanding regarding how teeth develop and how stem cells can be programmed. As a result, the possibilities of growing new teeth and of tooth bioengineering have been explored. Currently, a great deal is known about how signaling molecules and genes regulate tooth development, and modern research using transgenic mouse models has demonstrated that it is possible to induce the formation of new teeth by tinkering with the signaling networks that govern early tooth development. A breakthrough in stem cell biology in 2006 opened up the possibility that a patient's own cells can be programmed to develop into pluripotent stem cells and used for building new tissues and organs. At present, active research in numerous laboratories around the world addresses the question of how to program the stem and progenitor cells to develop into tooth-specific cell types. Taken together, the remarkable progress in developmental and stem cell biology is now feeding hopes of growing new teeth in the dental clinic in the not-too-distant future.
Collapse
Affiliation(s)
- Irma Thesleff
- Developmental Biology Research Program, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
42
|
Iop L, Assunta F, Gerosa G. Mechanical Circulatory Support and Stem Cell-Based Heart Treatment in Europe-2018 Clinical Update. Artif Organs 2019; 42:871-878. [PMID: 30328625 DOI: 10.1111/aor.13341] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 07/25/2018] [Accepted: 07/27/2018] [Indexed: 12/25/2022]
Affiliation(s)
- Laura Iop
- Cardiovascular Regenerative Medicine, Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Padua, Italy.,Venetian Institute of Molecular Medicine, Padua, Italy
| | - Fabozzo Assunta
- Cardiac Surgery, University Hospital of Padua, Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Padua, Italy
| | - Gino Gerosa
- Cardiovascular Regenerative Medicine, Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Padua, Italy.,Venetian Institute of Molecular Medicine, Padua, Italy.,Cardiac Surgery, University Hospital of Padua, Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Padua, Italy
| |
Collapse
|
43
|
Pranke I, Golec A, Hinzpeter A, Edelman A, Sermet-Gaudelus I. Emerging Therapeutic Approaches for Cystic Fibrosis. From Gene Editing to Personalized Medicine. Front Pharmacol 2019; 10:121. [PMID: 30873022 PMCID: PMC6400831 DOI: 10.3389/fphar.2019.00121] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 01/31/2019] [Indexed: 12/13/2022] Open
Abstract
An improved understanding of the cystic fibrosis (CF) transmembrane conductance regulator (CFTR) protein structure and the consequences of CFTR gene mutations have allowed the development of novel therapies targeting specific defects underlying CF. Some strategies are mutation specific and have already reached clinical development; some strategies include a read-through of the specific premature termination codons (read-through therapies, nonsense mediated decay pathway inhibitors for Class I mutations); correction of CFTR folding and trafficking to the apical plasma membrane (correctors for Class II mutations); and an increase in the function of CFTR channel (potentiators therapy for Class III mutations and any mutant with a residual function located at the membrane). Other therapies that are in preclinical development are not mutation specific and include gene therapy to edit the genome and stem cell therapy to repair the airway tissue. These strategies that are directed at the basic CF defects are now revolutionizing the treatment for patients and should positively impact their survival rates.
Collapse
Affiliation(s)
- Iwona Pranke
- INSERM U 1151, Institut Necker Enfants Malades, Université Paris Descartes, Paris, France
| | - Anita Golec
- INSERM U 1151, Institut Necker Enfants Malades, Université Paris Descartes, Paris, France
| | - Alexandre Hinzpeter
- INSERM U 1151, Institut Necker Enfants Malades, Université Paris Descartes, Paris, France
| | - Aleksander Edelman
- INSERM U 1151, Institut Necker Enfants Malades, Université Paris Descartes, Paris, France
| | - Isabelle Sermet-Gaudelus
- INSERM U 1151, Institut Necker Enfants Malades, Université Paris Descartes, Paris, France.,Centre de Référence Maladie Rare, Mucoviscidose et Maladies de CFTR, Paris, France.,Faculté de Médecine, Université Paris Descartes, Paris, France
| |
Collapse
|
44
|
Lei W, Li W, Ge L, Chen G. Non-engineered and Engineered Adult Neurogenesis in Mammalian Brains. Front Neurosci 2019; 13:131. [PMID: 30872991 PMCID: PMC6401632 DOI: 10.3389/fnins.2019.00131] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 02/05/2019] [Indexed: 12/31/2022] Open
Abstract
Adult neurogenesis has been extensively studied in rodent animals, with distinct niches found in the hippocampus and subventricular zone (SVZ). In non-human primates and human postmortem samples, there has been heated debate regarding adult neurogenesis, but it is largely agreed that the rate of adult neurogenesis is much reduced comparing to rodents. The limited adult neurogenesis may partly explain why human brains do not have self-repair capability after injury or disease. A new technology called “in vivo cell conversion” has been invented to convert brain internal glial cells in the injury areas directly into functional new neurons to replenish the lost neurons. Because glial cells are abundant throughout the brain and spinal cord, such engineered glia-to-neuron conversion technology can be applied throughout the central nervous system (CNS) to regenerate new neurons. Thus, compared to cell transplantation or the non-engineered adult neurogenesis, in vivo engineered neuroregeneration technology can provide a large number of functional new neurons in situ to repair damaged brain and spinal cord.
Collapse
Affiliation(s)
- Wenliang Lei
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Wen Li
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Longjiao Ge
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Gong Chen
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China.,Department of Biology, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, United States
| |
Collapse
|
45
|
Dolatshad H, Tatwavedi D, Ahmed D, Tegethoff JF, Boultwood J, Pellagatti A. Application of induced pluripotent stem cell technology for the investigation of hematological disorders. Adv Biol Regul 2019; 71:19-33. [PMID: 30341008 DOI: 10.1016/j.jbior.2018.10.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 10/08/2018] [Accepted: 10/09/2018] [Indexed: 06/08/2023]
Abstract
Induced pluripotent stem cells (iPSCs) were first described over a decade ago and are currently used in various basic biology and clinical research fields. Recent advances in the field of human iPSCs have opened the way to a better understanding of the biology of human diseases. Disease-specific iPSCs provide an unparalleled opportunity to establish novel human cell-based disease models, with the potential to enhance our understanding of the molecular mechanisms underlying human malignancies, and to accelerate the identification of effective new drugs. When combined with genome editing technologies, iPSCs represent a new approach to study single or multiple disease-causing mutations and model specific diseases in vitro. In addition, genetically corrected patient-specific iPSCs could potentially be used for stem cell based therapy. Furthermore, the reprogrammed cells share patient-specific genetic background, offering a new platform to develop personalized therapy/medicine for patients. In this review we discuss the recent advances in iPSC research technology and their potential applications in hematological diseases. Somatic cell reprogramming has presented new routes for generating patient-derived iPSCs, which can be differentiated to hematopoietic stem cells and the various downstream hematopoietic lineages. iPSC technology shows promise in the modeling of both inherited and acquired hematological disorders. A direct reprogramming and differentiation strategy is able to recapitulate hematological disorder progression and capture the earliest molecular alterations that underlie the initiation of hematological malignancies.
Collapse
Affiliation(s)
- Hamid Dolatshad
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, and Oxford BRC Haematology Theme, Oxford, UK
| | - Dharamveer Tatwavedi
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, and Oxford BRC Haematology Theme, Oxford, UK
| | - Doaa Ahmed
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, and Oxford BRC Haematology Theme, Oxford, UK; Clinical Pathology Department, Assiut University Hospitals, Faculty of Medicine, Assiut, Egypt
| | - Jana F Tegethoff
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, and Oxford BRC Haematology Theme, Oxford, UK
| | - Jacqueline Boultwood
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, and Oxford BRC Haematology Theme, Oxford, UK
| | - Andrea Pellagatti
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, and Oxford BRC Haematology Theme, Oxford, UK.
| |
Collapse
|
46
|
Affiliation(s)
- Kapil Bharti
- Ocular Stem Cell and Translational Research Unit at the National Eye Institute, NIH, Bethesda, Maryland, USA
| |
Collapse
|
47
|
Pigeau GM, Csaszar E, Dulgar-Tulloch A. Commercial Scale Manufacturing of Allogeneic Cell Therapy. Front Med (Lausanne) 2018; 5:233. [PMID: 30186836 PMCID: PMC6113399 DOI: 10.3389/fmed.2018.00233] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 07/31/2018] [Indexed: 12/14/2022] Open
Abstract
Allogeneic cell therapy products are generating encouraging clinical and pre-clinical results. Pluripotent stem cell (PSC) derived therapies, in particular, have substantial momentum and the potential to serve as treatments for a wide range of indications. Many of these therapies are also expected to have large market sizes and require cell doses of ≥109 cells. As therapeutic technologies mature, it is essential for the cell manufacturing industry to correspondingly develop to adequately support commercial scale production. To that end, there is much that can be learned and adapted from traditional manufacturing fields. In this review, we highlight key areas of allogeneic cell therapy manufacturing, identify current gaps, and discuss strategies for integrating new solutions. It is anticipated that cell therapy scale-up manufacturing solutions will need to generate batches of up to 2,000 L in single-use disposable formats, which constrains selection of currently available upstream hardware. Suitable downstream hardware is even more limited as processing solutions from the biopharmaceutical field are often not compatible with the unique requirements of cell therapy products. The advancement of therapeutic cell manufacturing processes to date has largely been developed with a cell biology driven approach, which is essential in early development. However, for truly robust and standardized production in a maturing field, a highly controlled manufacturing engineering strategy must be employed, with the implementation of automation, process monitoring and control to increase batch consistency and efficiency.
Collapse
Affiliation(s)
- Gary M Pigeau
- Centre for Advanced Therapeutic Cell Technologies, Toronto, ON, Canada.,General Electric Healthcare, Cell Therapy Technologies, Marlborough, MA, United States
| | - Elizabeth Csaszar
- Centre for Advanced Therapeutic Cell Technologies, Toronto, ON, Canada.,Centre for Commercialization of Regenerative Medicine, Toronto, ON, Canada
| | - Aaron Dulgar-Tulloch
- Centre for Advanced Therapeutic Cell Technologies, Toronto, ON, Canada.,General Electric Healthcare, Cell Therapy Technologies, Marlborough, MA, United States
| |
Collapse
|
48
|
Morena F, Argentati C, Bazzucchi M, Emiliani C, Martino S. Above the Epitranscriptome: RNA Modifications and Stem Cell Identity. Genes (Basel) 2018; 9:E329. [PMID: 29958477 PMCID: PMC6070936 DOI: 10.3390/genes9070329] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 06/15/2018] [Accepted: 06/25/2018] [Indexed: 02/07/2023] Open
Abstract
Sequence databases and transcriptome-wide mapping have revealed different reversible and dynamic chemical modifications of the nitrogen bases of RNA molecules. Modifications occur in coding RNAs and noncoding-RNAs post-transcriptionally and they can influence the RNA structure, metabolism, and function. The result is the expansion of the variety of the transcriptome. In fact, depending on the type of modification, RNA molecules enter into a specific program exerting the role of the player or/and the target in biological and pathological processes. Many research groups are exploring the role of RNA modifications (alias epitranscriptome) in cell proliferation, survival, and in more specialized activities. More recently, the role of RNA modifications has been also explored in stem cell biology. Our understanding in this context is still in its infancy. Available evidence addresses the role of RNA modifications in self-renewal, commitment, and differentiation processes of stem cells. In this review, we will focus on five epitranscriptomic marks: N6-methyladenosine, N1-methyladenosine, 5-methylcytosine, Pseudouridine (Ψ) and Adenosine-to-Inosine editing. We will provide insights into the function and the distribution of these chemical modifications in coding RNAs and noncoding-RNAs. Mainly, we will emphasize the role of epitranscriptomic mechanisms in the biology of naïve, primed, embryonic, adult, and cancer stem cells.
Collapse
Affiliation(s)
- Francesco Morena
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, 06126 Perugia, Italy.
| | - Chiara Argentati
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, 06126 Perugia, Italy.
| | - Martina Bazzucchi
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, 06126 Perugia, Italy.
| | - Carla Emiliani
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, 06126 Perugia, Italy.
- CEMIN, Center of Excellence of Nanostructured Innovative Materials, University of Perugia, 06126 Perugia, Italy.
| | - Sabata Martino
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, 06126 Perugia, Italy.
- CEMIN, Center of Excellence of Nanostructured Innovative Materials, University of Perugia, 06126 Perugia, Italy.
| |
Collapse
|
49
|
|
50
|
Singh LP. Gene and Tissue Engineering For the Treatment of Diabetes and Its Retinal Complications: The Use of Nucleic Acid Constructs Bearing A TXNIP Gene Promoter. CURRENT TRENDS IN BIOMEDICAL ENGINEERING & BIOSCIENCES 2018; 13. [PMID: 31355358 PMCID: PMC6660148 DOI: 10.19080/ctbeb.2018.13.555869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Diabetes is a chronic disease in which insulin production is deficient (Type 1) or resistant (Type 2) leading to organ complications including the heart, kidney, retina, and peripheral nerves. About 10% of diabetics are Type 1 while ~90 percent are Type 2 associated with life style changes and obesity. Whether it is Type 1 or Type 2, chronic hyperglycemia prevails and associated oxidative stress and low grade inflammation are considered to play critical roles in diabetes and its complications including diabetic retinopathy (DR). Thioredoxin-Interacting Protein, TXNIP, is strongly induced by diabetes and high glucose in all tissues examined including the pancreatic beta cells and the retina. TXNIP binds to and inhibits the anti-oxidant and thiol reducing capacity of thioredoxins and causes cellular oxidative stress, inflammation and premature cell death. TXNIP is induced strongly by high glucose and its metabolites with minutes and remains elevated as long as hyperglycemia persists. Therefore, the TXNIP gene promoter linked with insulin or a gene of interest may be used to induce gene expression or suppression and in tissue engineering for adipose or tissue-derived autologous stem cells producing insulin for the treatment of diabetes and its complications such as DR as well as age-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Lalit P Singh
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, USA.,Department of Ophthalmology, Wayne State University School of Medicine, USA
| |
Collapse
|