801
|
Balakrishnan A, Belfiore L, Chu TH, Fleming T, Midha R, Biernaskie J, Schuurmans C. Insights Into the Role and Potential of Schwann Cells for Peripheral Nerve Repair From Studies of Development and Injury. Front Mol Neurosci 2021; 13:608442. [PMID: 33568974 PMCID: PMC7868393 DOI: 10.3389/fnmol.2020.608442] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 12/31/2020] [Indexed: 12/13/2022] Open
Abstract
Peripheral nerve injuries arising from trauma or disease can lead to sensory and motor deficits and neuropathic pain. Despite the purported ability of the peripheral nerve to self-repair, lifelong disability is common. New molecular and cellular insights have begun to reveal why the peripheral nerve has limited repair capacity. The peripheral nerve is primarily comprised of axons and Schwann cells, the supporting glial cells that produce myelin to facilitate the rapid conduction of electrical impulses. Schwann cells are required for successful nerve regeneration; they partially “de-differentiate” in response to injury, re-initiating the expression of developmental genes that support nerve repair. However, Schwann cell dysfunction, which occurs in chronic nerve injury, disease, and aging, limits their capacity to support endogenous repair, worsening patient outcomes. Cell replacement-based therapeutic approaches using exogenous Schwann cells could be curative, but not all Schwann cells have a “repair” phenotype, defined as the ability to promote axonal growth, maintain a proliferative phenotype, and remyelinate axons. Two cell replacement strategies are being championed for peripheral nerve repair: prospective isolation of “repair” Schwann cells for autologous cell transplants, which is hampered by supply challenges, and directed differentiation of pluripotent stem cells or lineage conversion of accessible somatic cells to induced Schwann cells, with the potential of “unlimited” supply. All approaches require a solid understanding of the molecular mechanisms guiding Schwann cell development and the repair phenotype, which we review herein. Together these studies provide essential context for current efforts to design glial cell-based therapies for peripheral nerve regeneration.
Collapse
Affiliation(s)
- Anjali Balakrishnan
- Biological Sciences Platform, Sunnybrook Research Institute (SRI), Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Lauren Belfiore
- Biological Sciences Platform, Sunnybrook Research Institute (SRI), Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Tak-Ho Chu
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Taylor Fleming
- Biological Sciences Platform, Sunnybrook Research Institute (SRI), Toronto, ON, Canada
| | - Rajiv Midha
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Jeff Biernaskie
- Department of Comparative Biology and Experimental Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Carol Schuurmans
- Biological Sciences Platform, Sunnybrook Research Institute (SRI), Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
802
|
Davim A, Trindade da silva L, Vieira P. Environmental Enrichment as a Strategy to Confront Social Isolation Under the COVID-19 Pandemic. Front Behav Neurosci 2021; 14:564184. [PMID: 33551762 PMCID: PMC7859510 DOI: 10.3389/fnbeh.2020.564184] [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: 05/20/2020] [Accepted: 11/30/2020] [Indexed: 12/20/2022] Open
Abstract
The moment of social isolation experienced by the world population due to the COVID-19 pandemic tends to trigger behavioral changes of different orders and on an exponential scale, regardless of social class, age, gender, or ethnicity. Environmental enrichment presents itself as an important strategy to face the social isolation imposed by the pandemic, in order to act as an important agent of induction of biological factors for cognitive and emotional development, favoring a better possibility of adaptation to isolation.
Collapse
Affiliation(s)
- André Davim
- Departament of Morphology, Centro Universitário do Rio Grande do Norte, Natal, Brazil
- Febracis Institution, Fortaleza, Brazil
| | | | | |
Collapse
|
803
|
Azizipour E, Aghamollaei H, Halabian R, Poormoghadam D, Saffari M, Entezari M, Salimi A. A novel hydrogel scaffold contained bioactive glass nanowhisker (BGnW) for osteogenic differentiation of human mesenchymal stem cells (hMSCs) in vitro. Int J Biol Macromol 2021; 174:562-572. [PMID: 33434552 DOI: 10.1016/j.ijbiomac.2021.01.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/26/2020] [Accepted: 01/01/2021] [Indexed: 12/18/2022]
Abstract
Employing hydrogels as an alternative strategy for repairing bone defects has received great attention in bone tissue engineering. In this study, hydrogel scaffold based on collagen, gelatin, and glutaraldehyde was combined with bioactive glass nanowhiskers (BGnW) to differentiate human mesenchymal stem cells (hMSCs) into the osteogenic lineage and inducing biomineralization. Pure Gel-Glu-Col and bioactive glass nanowhiskers were used as control throughout the paper. Chemical, physical and morphological characteristics of the nanocomposite scaffold were assessed meticulously using Fourier transform infrared spectroscopy (FTIR), X-ray diffraction (XRD), porosity measurement, water uptake ability, tensile test, and scanning electron microscopy (SEM). To determine the cytotoxicity and cell viability of the hydrogel, MTT assay and Acridine orange (AO) staining were performed. hMSCs seeded on Gel-Glu-Col/BGnW were then incubated with osteogenic differentiation media for 14 days. Biomineralization assays (alkaline phosphatase (ALP) activity, calcium content assay, von Kossa, and Alizarin red staining) were carried out, and osteogenic genes and protein markers were examined using real time-PCR and immunocytochemistry. Results showed that the components of the hydrogel were properly integrated. The mechanical property of hydrogel was enhanced following the addition of BGnW. Cell viability assays confirmed the biocompatibility of the scaffold and increasing the proliferation after incorporating BGnW into pure Ge1-Glu-Col. Our nanocomposite maintained an enhanced ability of biomineralization as compared to its pure counterparts. Molecular investigations revealed an elevated level of osteogenic markers as compared to Ge1-Glu-Col and BGnW. All in all, Gel-Glu-Col/BGnW seems to be a potential candidate for the regeneration of bone tissue.
Collapse
Affiliation(s)
- Esmat Azizipour
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hossein Aghamollaei
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Raheleh Halabian
- Applied Microbiology Research Center, Baqiyatallah University Medical of Sciences, Tehran, Iran
| | - Delaram Poormoghadam
- Department of Medical Nanotechnology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mostafa Saffari
- Department of Pharmaceutics, School of Pharmacy, Islamic Azad University of Medical Sciences, Tehran, Iran
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Ali Salimi
- Nanobiotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
804
|
Transplanted embryonic retinal stem cells have the potential to repair the injured retina in mice. BMC Ophthalmol 2021; 21:26. [PMID: 33422026 PMCID: PMC7797095 DOI: 10.1186/s12886-020-01795-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 12/26/2020] [Indexed: 01/25/2023] Open
Abstract
Background Stem cell transplantation has been reported as one of the promising strategies to treat retinal degenerative diseases. But, the application and the role of retina stem cells (RSCs) in the treatment of patients with retinal degenerative diseases have not been fully revealed. This study aimed to investigate the potential role of transplantation of the embryo-derived RSCs into the vitreous cavity in repairing the damaged retina in mice. Methods RSCs were isolated from Kunming mice E17 embryonic retina and ciliary body tissues, and labeled with 5-bromo-2’-deoxyuridin (BrdU). Retinal optic nerve crush injury was induced in left eyes in male Kunming mice by ring clamping the optic nerve. The 6th -generation of BrdU-labeled RSCs were transplanted into the damaged retina by the intravitreal injection, and saline injected eyes were used as the control. Hematoxylin and eosin histological staining, and BrdU, Nestin and Pax6 immunostaining were performed. Electroretinogram (ERG) was used for assessing the electrical activity of the retina. Results Embryo-derived RSCs were identified by the positive stains of Pax6 and Nestin. BrdU incorporation was detected in the majority of RSCs. The damaged retina showed cellular nuclear disintegration and fragmentation in the retinal tissue which progressed over the periods of clamping time, and decreased amplitudes of a and b waves in ERG. In the damaged retina with RSCs transplantation, the positive staining for BrdU, Pax6 and Nestin were revealed on the retinal surface. Notably, RSCs migrated into the retinal ganglion cell layer and inner nuclear. Transplanted RSCs significantly elevated the amplitudes of a waves in retina injured eyes. Conclusions Embryonic RSCs have similar characteristics to neural stem cells. Transplantation of RSCs by intravitreal injection would be able to repair the damaged retina.
Collapse
|
805
|
Bagheri HS, Karimipour M, Heidarzadeh M, Rajabi H, Sokullu E, Rahbarghazi R. Does the Global Outbreak of COVID-19 or Other Viral Diseases Threaten the Stem Cell Reservoir Inside the Body? Stem Cell Rev Rep 2021; 17:214-230. [PMID: 33403490 PMCID: PMC7785129 DOI: 10.1007/s12015-020-10108-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2020] [Indexed: 12/20/2022]
Abstract
The COVID-19 pandemic has profoundly influenced public health and contributed to global economic divergences of unprecedented dimensions. Due to the high prevalence and mortality rates, it is then expected that the consequence and public health challenges will last for long periods. The rapid global spread of COVID-19 and lack of enough data regarding the virus pathogenicity multiplies the complexity and forced governments to react quickly against this pandemic. Stem cells represent a small fraction of cells located in different tissues. These cells play a critical role in the regeneration and restoration of injured sites. Because of their specific niche and a limited number of stem cells, the key question is whether there are different anti-viral mechanisms against viral infection notably COVID-19. Here, we aimed to highlight the intrinsic antiviral resistance in different stem cells against viral infection. These data could help us to understand the possible viral infections in different stem cells and the activation of specific molecular mechanisms upon viral entrance.
Collapse
Affiliation(s)
| | - Mohammad Karimipour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Heidarzadeh
- Koç University Translational Medicine Research Center (KUTTAM) Rumeli Feneri, Sarıyer, Istanbul, Turkey
| | - Hadi Rajabi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Emel Sokullu
- Koç University Translational Medicine Research Center (KUTTAM) Rumeli Feneri, Sarıyer, Istanbul, Turkey. .,School of Medicine, Biophysics Department, Koç University, Rumeli Fener, Sarıyer, Istanbul, Turkey.
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
806
|
Lee H, Song Y, Park YH, Uddin MS, Park JB. Evaluation of the Effects of Cuminum cyminum on Cellular Viability, Osteogenic Differentiation and Mineralization of Human Bone Marrow-Derived Stem Cells. ACTA ACUST UNITED AC 2021; 57:medicina57010038. [PMID: 33406654 PMCID: PMC7823674 DOI: 10.3390/medicina57010038] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 12/28/2020] [Accepted: 12/30/2020] [Indexed: 02/06/2023]
Abstract
Background and Objectives: Cuminum cyminum L. has long been used in the treatment of various diseases in multiple geographical regions. This study was performed to determine the effects of C. cyminum methanolic extract (CCT) on the cellular viability, alkaline phosphatase activity and mineralization of human mesenchymal stem cells. Materials and Methods: Bone marrow-derived stem cells were cultured in the presence of CCT at concentrations of 0, 0.001, 0.01, 0.1 and 1 μg/mL. Evaluations of cell morphology were performed on days 1, 3, 7 and 14. Cellular viability was evaluated on days 1, 3, 5 and 7. On the 7th and 14th day, alkaline phosphatase activity measurements and Alizarin red S staining were conducted to assess the osteogenic differentiation of stem cells. A real-time polymerase chain reaction was used to determine the expression levels of RUNX2, BSP, OCN, COL2A1 and β-catenin mRNAs. Results: Stem cells in the control group showed fibroblast-like morphology and the addition of CCT at 0.001, 0.01, 0.1 and 1 μg/mL did not generate noticeable changes in morphology compared with the untreated control group. The application of CCT did not produce significant changes in cellular viability or alkaline phosphatase activity compared with controls. Alizarin Red S staining was significantly increased with the application of CCT. Treatment with CCT increased the expressions of RUNX2, BSP and OCN. Conclusions: These results indicate that CCT enhanced the osteogenic differentiation of stem cells derived from bone marrow by regulating the expressions of RUNX2, BSP and OCN. Thus, the use of CCT may be applied to achieve beneficial effects on the mineralization of stem cells.
Collapse
Affiliation(s)
- Hyunjin Lee
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (H.L.); (Y.S.)
| | - Youngmin Song
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (H.L.); (Y.S.)
| | | | - Md. Salah Uddin
- Ethnobotanical Database of Bangladesh, Tejgaon, Dhaka 1208, Bangladesh;
| | - Jun-Beom Park
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (H.L.); (Y.S.)
- Correspondence: ; Tel.: +82-2-2258-6290
| |
Collapse
|
807
|
Pacheco-Herrero M, Soto-Rojas LO, Reyes-Sabater H, Garcés-Ramirez L, de la Cruz López F, Villanueva-Fierro I, Luna-Muñoz J. Current Status and Challenges of Stem Cell Treatment for Alzheimer's Disease. J Alzheimers Dis 2021; 84:917-935. [PMID: 34633316 PMCID: PMC8673502 DOI: 10.3233/jad-200863] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2021] [Indexed: 12/23/2022]
Abstract
Neurodegenerative diseases called tauopathies, such as Alzheimer's disease (AD), frontotemporal dementia, progressive supranuclear palsy, and Parkinson's disease, among others, are characterized by the pathological processing and accumulation of tau protein. AD is the most prevalent neurodegenerative disease and is characterized by two lesions: neurofibrillary tangles (NFTs) and neuritic plaques. The presence of NFTs in the hippocampus and neocortex in early and advanced stages, respectively, correlates with the patient's cognitive deterioration. So far, no drugs can prevent, decrease, or limit neuronal death due to abnormal pathological tau accumulation. Among potential non-pharmacological treatments, physical exercise has been shown to stimulate the development of stem cells (SCs) and may be useful in early stages. However, this does not prevent neuronal death from the massive accumulation of NFTs. In recent years, SCs therapies have emerged as a promising tool to repopulate areas involved in cognition in neurodegenerative diseases. Unfortunately, protocols for SCs therapy are still being developed and the mechanism of action of such therapy remains unclear. In this review, we show the advances and limitations of SCs therapy. Finally, we provide a critical analysis of its clinical use for AD.
Collapse
Affiliation(s)
- Mar Pacheco-Herrero
- Neuroscience Research Laboratory, Faculty of Health Sciences, Pontificia Universidad Católica Madre y Maestra, Dominican Republic
| | - Luis O. Soto-Rojas
- Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, State of Mexico, Mexico
| | - Heidy Reyes-Sabater
- Neuroscience Research Laboratory, Faculty of Health Sciences, Pontificia Universidad Católica Madre y Maestra, Dominican Republic
| | - Linda Garcés-Ramirez
- Escuela Nacional de Ciencias Biológicas, Depto de Fisiología, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Fidel de la Cruz López
- Escuela Nacional de Ciencias Biológicas, Depto de Fisiología, Instituto Politécnico Nacional, Mexico City, Mexico
| | | | - José Luna-Muñoz
- National Dementia BioBank, Ciencias Biológicas, Facultad de Estudios Superiores Cuautitlán, UNAM, State of Mexico, Mexico
- Banco Nacional de Cerebros-UNPHU, Universidad Nacional Pedro Henríquez Ureña, Dominican Republic
| |
Collapse
|
808
|
Stem Cells an Overview. Stem Cells 2021. [DOI: 10.1007/978-981-16-1638-9_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
809
|
Küçükgüven MB, Çelebi-Saltik B. Stem Cell Based Exosomes: Are They Effective in Disease or Health? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1341:45-65. [PMID: 33782904 DOI: 10.1007/5584_2021_630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Exosomes are nano-sized vesicles involved in intercellular communication via delivery of molecules including lipids, nucleic acids, proteins, or other cellular components to distant or neighboring sites. Their ability to pass biological barriers, stability in physiological fluids without degradation, and distinctive affinity to target cells make exosomes very remarkable therapeutic vehicles. Virus-based approaches are some of the most widely used gene therapy methods; however, there are many issues need to be clarified such as high immunogenicity. Using of the exosomes procures the functional transfer of their cargo with minimal intervention from the immune system and it has been reported to be secure and well-tolerated. When the regenerative medicine is taken into consideration, stem cell-based approaches have been aimed to utilize but the general efficacy and safety profile of stem cell therapy has still not been enlightened. At this point, stem cell-derived exosomes exhibit a way to procure cell-free regenerative medicine with their unique characteristics. Exosomes are considered as appropriate and highly stable biological nano-vectors taking part in a wide variety of healthy and pathological processes for advanced targeted therapies. However, there are still crucial obstacles to achieve efficient isolation of large amount of specific and pure exosomes. Thus, large-scale exosome production under good manufacturing practice is required. The purpose of this review is to focus on stem cell-based exosomes for gene delivery and to introduce synthetic exosome-mimics as a potential alternative in the field of targeted gene therapies. Further, we aim to highlight the biobanking and large-scale manufacturing methods of exosomes.
Collapse
Affiliation(s)
- Meriç Bilgiç Küçükgüven
- Department of Oral and Maxillofacial Surgery, Hacettepe University Faculty of Dentistry, Ankara, Turkey.,Department of Stem Cell Sciences, Hacettepe University Graduate School of Health Sciences, Ankara, Turkey.,Center for Stem Cell Research and Development, Hacettepe University, Ankara, Turkey
| | - Betül Çelebi-Saltik
- Department of Stem Cell Sciences, Hacettepe University Graduate School of Health Sciences, Ankara, Turkey. .,Center for Stem Cell Research and Development, Hacettepe University, Ankara, Turkey.
| |
Collapse
|
810
|
Jensen T, Wanczyk H, Thaker S, Finck C. Characterization of mesenchymal stem cells in patients with esophageal atresia. J Pediatr Surg 2021; 56:17-25. [PMID: 33121738 DOI: 10.1016/j.jpedsurg.2020.09.039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/14/2020] [Accepted: 09/15/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND Preclinical studies demonstrate that tissue engineering and patient-derived stem cells can regenerate tissue. The goal of this study was to determine whether stem cells from esophageal atresia patients (EA) could be utilized for this purpose. METHODS Adipose tissue was obtained from control, esophageal atresia (EA) and long gap esophageal atresia (LGEA) patients. Mesenchymal stem cells (MSCs) were isolated, expanded, characterized and seeded onto tubular scaffolds for 6 days. Scaffolds were characterized for viability, gene expression and cytokine production. RESULTS The average weight of tissue from the EA and LGEA patients was 145.8mg compared to 2981 mg in controls. Despite the small amount of tissue obtained from neonatal patients, cells were expanded to cover a scaffold. After incubating 6 days on the scaffold, cells were viable and proliferating with differences in gene expression between groups. VEGFA production in the supernatant was increased in EA and LGEA patients; while IL6 production was significantly increased in the control patients. CONCLUSIONS This study demonstrates the ability to utilize small amounts of adipose tissue from esophageal atresia patients as a cell source for regenerative medicine. Future studies will focus on use of these cells for tissue regeneration in vivo.
Collapse
Affiliation(s)
- Todd Jensen
- University of Connecticut School of Medicine, Department of Pediatrics, Farmington, CT.
| | - Heather Wanczyk
- University of Connecticut School of Medicine, Department of Pediatrics, Farmington, CT
| | | | - Christine Finck
- University of Connecticut School of Medicine, Department of Pediatrics, Farmington, CT; CT Children's, Department of Pediatric Surgery, Hartford, CT.
| |
Collapse
|
811
|
Jia TZ, Wang PH, Niwa T, Mamajanov I. Connecting primitive phase separation to biotechnology, synthetic biology, and engineering. J Biosci 2021; 46:79. [PMID: 34373367 PMCID: PMC8342986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 07/20/2021] [Indexed: 02/07/2023]
Abstract
One aspect of the study of the origins of life focuses on how primitive chemistries assembled into the first cells on Earth and how these primitive cells evolved into modern cells. Membraneless droplets generated from liquid-liquid phase separation (LLPS) are one potential primitive cell-like compartment; current research in origins of life includes study of the structure, function, and evolution of such systems. However, the goal of primitive LLPS research is not simply curiosity or striving to understand one of life's biggest unanswered questions, but also the possibility to discover functions or structures useful for application in the modern day. Many applicational fields, including biotechnology, synthetic biology, and engineering, utilize similar phaseseparated structures to accomplish specific functions afforded by LLPS. Here, we briefly review LLPS applied to primitive compartment research and then present some examples of LLPS applied to biomolecule purification, drug delivery, artificial cell construction, waste and pollution management, and flavor encapsulation. Due to a significant focus on similar functions and structures, there appears to be much for origins of life researchers to learn from those working on LLPS in applicational fields, and vice versa, and we hope that such researchers can start meaningful cross-disciplinary collaborations in the future.
Collapse
Affiliation(s)
- Tony Z Jia
- Earth-Life Science Institute, Tokyo Institute of Technology, 2-12-1-IE-1 Ookayama, Meguro-ku, Tokyo, 152-8550 Japan
- Blue Marble Space Institute of Science, 1001 4th Ave., Suite 3201, Seattle, Washington 98154 USA
| | - Po-Hsiang Wang
- Earth-Life Science Institute, Tokyo Institute of Technology, 2-12-1-IE-1 Ookayama, Meguro-ku, Tokyo, 152-8550 Japan
- Graduate Institute of Environmental Engineering, National Central University, Zhongli Dist, 300 Zhongda Rd, Taoyuan City, 32001 Taiwan
| | - Tatsuya Niwa
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Nagatsuta-cho 4259, Midori-ku, Yokohama, 226-8503 Japan
| | - Irena Mamajanov
- Earth-Life Science Institute, Tokyo Institute of Technology, 2-12-1-IE-1 Ookayama, Meguro-ku, Tokyo, 152-8550 Japan
| |
Collapse
|
812
|
Chia WK, Cheah FC, Abdul Aziz NH, Kampan NC, Shuib S, Khong TY, Tan GC, Wong YP. A Review of Placenta and Umbilical Cord-Derived Stem Cells and the Immunomodulatory Basis of Their Therapeutic Potential in Bronchopulmonary Dysplasia. Front Pediatr 2021; 9:615508. [PMID: 33791258 PMCID: PMC8006350 DOI: 10.3389/fped.2021.615508] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 02/17/2021] [Indexed: 12/13/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a devastating lung disorder of preterm infants as a result of an aberrant reparative response following exposures to various antenatal and postnatal insults. Despite sophisticated medical treatment in this modern era, the incidence of BPD remains unabated. The current strategies to prevent and treat BPD have met with limited success. The emergence of stem cell therapy may be a potential breakthrough in mitigating this complex chronic lung disorder. Over the last two decades, the human placenta and umbilical cord have gained increasing attention as a highly potential source of stem cells. Placenta-derived stem cells (PDSCs) and umbilical cord-derived stem cells (UCDSCs) display several advantages such as immune tolerance and are generally devoid of ethical constraints, in addition to their stemness qualities. They possess the characteristics of both embryonic and mesenchymal stromal/stem cells. Recently, there are many preclinical studies investigating the use of these cells as therapeutic agents in neonatal disease models for clinical applications. In this review, we describe the preclinical and clinical studies using PDSCs and UCDSCs as treatment in animal models of BPD. The source of these stem cells, routes of administration, and effects on immunomodulation, inflammation and regeneration in the injured lung are also discussed. Lastly, a brief description summarized the completed and ongoing clinical trials using PDSCs and UCDSCs as therapeutic agents in preventing or treating BPD. Due to the complexity of BPD, the development of a safe and efficient therapeutic agent remains a major challenge to both clinicians and researchers.
Collapse
Affiliation(s)
- Wai Kit Chia
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Fook Choe Cheah
- Department of Pediatrics, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Nor Haslinda Abdul Aziz
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Nirmala Chandralega Kampan
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Salwati Shuib
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Teck Yee Khong
- Department of Pathology, SA Pathology, Women's and Children's Hospital, Adelaide, SA, Australia
| | - Geok Chin Tan
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Yin Ping Wong
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
813
|
Vasanthan J, Gurusamy N, Rajasingh S, Sigamani V, Kirankumar S, Thomas EL, Rajasingh J. Role of Human Mesenchymal Stem Cells in Regenerative Therapy. Cells 2020; 10:E54. [PMID: 33396426 PMCID: PMC7823630 DOI: 10.3390/cells10010054] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/27/2020] [Accepted: 12/29/2020] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells which can proliferate and replace dead cells in the body. MSCs also secrete immunomodulatory molecules, creating a regenerative microenvironment that has an excellent potential for tissue regeneration. MSCs can be easily isolated and grown in vitro for various applications. For the past two decades, MSCs have been used in research, and many assays and tests have been developed proving that MSCs are an excellent cell source for therapy. This review focusses on quality control parameters required for applications of MSCs including colony formation, surface markers, differentiation potentials, and telomere length. Further, the specific mechanisms of action of MSCs under various conditions such as trans-differentiation, cell fusion, mitochondrial transfer, and secretion of extracellular vesicles are discussed. This review aims to underline the applications and benefits of MSCs in regenerative medicine and tissue engineering.
Collapse
Affiliation(s)
- Jayavardini Vasanthan
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (J.V.); (N.G.); (S.R.); (V.S.); (S.K.); (E.L.T.)
- Department of Genetic Engineering, SRM Institute of Science and Technology, Chennai 600036, India
| | - Narasimman Gurusamy
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (J.V.); (N.G.); (S.R.); (V.S.); (S.K.); (E.L.T.)
| | - Sheeja Rajasingh
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (J.V.); (N.G.); (S.R.); (V.S.); (S.K.); (E.L.T.)
| | - Vinoth Sigamani
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (J.V.); (N.G.); (S.R.); (V.S.); (S.K.); (E.L.T.)
| | - Shivaani Kirankumar
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (J.V.); (N.G.); (S.R.); (V.S.); (S.K.); (E.L.T.)
- Department of Genetic Engineering, SRM Institute of Science and Technology, Chennai 600036, India
| | - Edwin L. Thomas
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (J.V.); (N.G.); (S.R.); (V.S.); (S.K.); (E.L.T.)
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Johnson Rajasingh
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (J.V.); (N.G.); (S.R.); (V.S.); (S.K.); (E.L.T.)
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
814
|
Kokkorakis N, Gaitanou M. Minibrain-related kinase/dual-specificity tyrosine-regulated kinase 1B implication in stem/cancer stem cells biology. World J Stem Cells 2020; 12:1553-1575. [PMID: 33505600 PMCID: PMC7789127 DOI: 10.4252/wjsc.v12.i12.1553] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/29/2020] [Accepted: 10/15/2020] [Indexed: 02/06/2023] Open
Abstract
Dual-specificity tyrosine phosphorylation-regulated kinase 1B (DYRK1B), also known as minibrain-related kinase (MIRK) is one of the best functionally studied members of the DYRK kinase family. DYRKs comprise a family of protein kinases that are emerging modulators of signal transduction pathways, cell proliferation and differentiation, survival, and cell motility. DYRKs were found to participate in several signaling pathways critical for development and cell homeostasis. In this review, we focus on the DYRK1B protein kinase from a functional point of view concerning the signaling pathways through which DYRK1B exerts its cell type-dependent function in a positive or negative manner, in development and human diseases. In particular, we focus on the physiological role of DYRK1B in behavior of stem cells in myogenesis, adipogenesis, spermatogenesis and neurogenesis, as well as in its pathological implication in cancer and metabolic syndrome. Thus, understanding of the molecular mechanisms that regulate signaling pathways is of high importance. Recent studies have identified a close regulatory connection between DYRK1B and the hedgehog (HH) signaling pathway. Here, we aim to bring together what is known about the functional integration and cross-talk between DYRK1B and several signaling pathways, such as HH, RAS and PI3K/mTOR/AKT, as well as how this might affect cellular and molecular processes in development, physiology, and pathology. Thus, this review summarizes the major known functions of DYRK1B kinase, as well as the mechanisms by which DYRK1B exerts its functions in development and human diseases focusing on the homeostasis of stem and cancer stem cells.
Collapse
Affiliation(s)
- Nikolaos Kokkorakis
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Hellenic Pasteur Institute, Athens 11521, Greece
| | - Maria Gaitanou
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Hellenic Pasteur Institute, Athens 11521, Greece.
| |
Collapse
|
815
|
Park JS, Park G, Hong HS. Age affects the paracrine activity and differentiation potential of human adipose‑derived stem cells. Mol Med Rep 2020; 23:160. [PMID: 33655325 PMCID: PMC7789087 DOI: 10.3892/mmr.2020.11799] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 12/03/2020] [Indexed: 01/09/2023] Open
Abstract
Stem cell therapy is considered a novel treatment modality for critical diseases. Adipose tissue is a rich and easily accessible source of stem cells. Adipose‑derived stem cells (ADSCs) can be expanded ex vivo and possess characteristics similar to those derived from the bone marrow. However, the quality of ADSCs can be affected by age, underlying disease or the lifestyle of individuals. The aim of the present study was to explore the association between age and ADSC activity, including paracrine and differentiation potential. Adipose tissues from young (age <30 years) and elderly (age >70 years) groups were obtained, and ADSCs from each group were cultured <em>in vitro</em>. The effect of age on ADSC activity was investigated <em>in vitro</em> by evaluating the proliferation rate, adipo/osteogenic differentiation potential and cytokine profile using ELISA. The results revealed that increased age reduced cell activity and increased the doubling time of ADSCs, without causing profound morphological changes. The paracrine action of ADSCs was markedly altered by increased age, as demonstrated by reduced expression levels of vascular endothelial growth factor, stromal cell‑derived factor‑1α and hepatocyte growth factor. Differentiation of ADSCs into osteoblasts or adipocytes rarely occurred in the elderly group compared with the young group. Overall, these results indicate that age may affect the cellular function of ADSCs and should be considered prior to ADSC transplantation.
Collapse
Affiliation(s)
- Jeong Seop Park
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Gabee Park
- Department of Genetic Engineering, College of Life Science and Graduate School of Biotechnology, Kyung Hee University, Yong In 17104, Republic of Korea
| | - Hyun Sook Hong
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
816
|
Kim K, Bou-Ghannam S, Kameishi S, Oka M, Grainger DW, Okano T. Allogeneic mesenchymal stem cell sheet therapy: A new frontier in drug delivery systems. J Control Release 2020; 330:696-704. [PMID: 33347942 DOI: 10.1016/j.jconrel.2020.12.028] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 12/13/2022]
Abstract
The evolution of drug discovery exploded in the early 20th century with the advent of critical scientific advancements in organic chemistry, chemical analysis, and purification. Early drug generations focused largely on symptom control and pain management, effective targets for small-molecule drugs. Recently, the attention in drug discovery has shifted to pursuit of radical cures. Cell therapy presents the ideal attributes of a promising new drug, targeting specific tissues based on chemotactic cues and modulating secretion of instructive regenerative molecules in response to dynamic signaling from disease environments. To actuate the therapeutic potential of cell therapy toward worldwide clinical use, cell delivery methods that can effectively localize and engraft mesenchymal stem cells (MSCs) with high disease-site fidelity and enable dynamic MSC bioactive function are paramount. In this review, we discuss the evolution of cell therapies with a focus on stem cell advantages, as well as the limitations to these therapies. This review aims to introduce cell sheet technology as a breakthrough cell therapy with demonstrated therapeutic success across indications for heart, liver, and kidney tissue regeneration. Opportunities and anticipated clinical impacts of cell sheet technology using MSCs are discussed.
Collapse
Affiliation(s)
- Kyungsook Kim
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, Health Sciences, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA.
| | - Sophia Bou-Ghannam
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, Health Sciences, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA; Department of Biomedical Engineering, University of Utah, 36 South, Wasatch Drive, Salt Lake City, UT 84112, USA
| | - Sumako Kameishi
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, Health Sciences, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA
| | - Masatoshi Oka
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, Health Sciences, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA
| | - David W Grainger
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, Health Sciences, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA; Department of Biomedical Engineering, University of Utah, 36 South, Wasatch Drive, Salt Lake City, UT 84112, USA
| | - Teruo Okano
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, Health Sciences, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA; Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
| |
Collapse
|
817
|
Aiyegbusi OL, Macpherson K, Elston L, Myles S, Washington J, Sungum N, Briggs M, Newsome PN, Calvert MJ. Patient and public perspectives on cell and gene therapies: a systematic review. Nat Commun 2020; 11:6265. [PMID: 33293538 PMCID: PMC7722871 DOI: 10.1038/s41467-020-20096-1] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 11/11/2020] [Indexed: 12/12/2022] Open
Abstract
Cell and gene therapies offer opportunities for treating disease with potential to restore function, and cure disease. However, they are not without risk and pose complex logistical, economic, ethical and social challenges for health systems. Here we report our systematic review of the current evidence on patient and public knowledge and perspectives of cell and gene therapies, to inform future research, education and awareness raising activities. We screened 10,735 titles and abstracts, and evaluated the full texts of 151 publications. The final selection was 35 publications. Four themes were generated from the narrative synthesis of the study findings namely: (1) Knowledge and understanding of cell and gene therapies, (2) Acceptance of cell and gene therapies (3) Understanding of risk and benefits of therapy, and (4) Information needs and current sources of information. As potential funders or future recipients, it is important that the public and patients are aware of these therapies, understand the issues involved, and can contribute to the debate. This review highlights the need for appropriate patient and public education on the various aspects of cell and gene therapies. High quality studies exploring patient and public opinions and experiences of cell and gene therapy are required. Patient and public perceptions of these therapies, alongside evidence of clinical and cost-effectiveness, will be central to their uptake and use.
Collapse
Affiliation(s)
- Olalekan Lee Aiyegbusi
- Centre for Patient Reported Outcomes Research, Institute of Applied Health Research, University of Birmingham, Birmingham, UK.
- National Institute for Health Research Birmingham Biomedical Research Centre, University of Birmingham, Birmingham, UK.
- National Institute for Health Research Applied Research Centre West Midlands, and National Institute for Health Research Surgical Reconstruction and Microbiology Research Centre, University of Birmingham, Birmingham, UK.
- Birmingham Health Partners Centre for Regulatory Science and Innovation, Birmingham, UK.
| | | | | | | | | | - Nisha Sungum
- Birmingham Health Partners Centre for Regulatory Science and Innovation, Birmingham, UK
- Midlands-Wales Advanced Therapy Treatment Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Mark Briggs
- Welsh Blood Service, Velindre University NHS Trust, Cardiff, UK
| | - Philip N Newsome
- National Institute for Health Research Birmingham Biomedical Research Centre, University of Birmingham, Birmingham, UK
- National Institute for Health Research Applied Research Centre West Midlands, and National Institute for Health Research Surgical Reconstruction and Microbiology Research Centre, University of Birmingham, Birmingham, UK
- Birmingham Health Partners Centre for Regulatory Science and Innovation, Birmingham, UK
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
- Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Melanie J Calvert
- Centre for Patient Reported Outcomes Research, Institute of Applied Health Research, University of Birmingham, Birmingham, UK
- National Institute for Health Research Birmingham Biomedical Research Centre, University of Birmingham, Birmingham, UK
- National Institute for Health Research Applied Research Centre West Midlands, and National Institute for Health Research Surgical Reconstruction and Microbiology Research Centre, University of Birmingham, Birmingham, UK
- Birmingham Health Partners Centre for Regulatory Science and Innovation, Birmingham, UK
| |
Collapse
|
818
|
Culibrk RA, Hahn MS. The Role of Chronic Inflammatory Bone and Joint Disorders in the Pathogenesis and Progression of Alzheimer's Disease. Front Aging Neurosci 2020; 12:583884. [PMID: 33364931 PMCID: PMC7750365 DOI: 10.3389/fnagi.2020.583884] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 11/06/2020] [Indexed: 12/12/2022] Open
Abstract
Late-onset Alzheimer's Disease (LOAD) is a devastating neurodegenerative disorder that causes significant cognitive debilitation in tens of millions of patients worldwide. Throughout disease progression, abnormal secretase activity results in the aberrant cleavage and subsequent aggregation of neurotoxic Aβ plaques in the cerebral extracellular space and hyperphosphorylation and destabilization of structural tau proteins surrounding neuronal microtubules. Both pathologies ultimately incite the propagation of a disease-associated subset of microglia-the principle immune cells of the brain-characterized by preferentially pro-inflammatory cytokine secretion and inhibited AD substrate uptake capacity, which further contribute to neuronal degeneration. For decades, chronic neuroinflammation has been identified as one of the cardinal pathophysiological driving features of AD; however, despite a number of works postulating the underlying mechanisms of inflammation-mediated neurodegeneration, its pathogenesis and relation to the inception of cognitive impairment remain obscure. Moreover, the limited clinical success of treatments targeting specific pathological features in the central nervous system (CNS) illustrates the need to investigate alternative, more holistic approaches for ameliorating AD outcomes. Accumulating evidence suggests significant interplay between peripheral immune activity and blood-brain barrier permeability, microglial activation and proliferation, and AD-related cognitive decline. In this work, we review a narrow but significant subset of chronic peripheral inflammatory conditions, describe how these pathologies are associated with the preponderance of neuroinflammation, and posit that we may exploit peripheral immune processes to design interventional, preventative therapies for LOAD. We then provide a comprehensive overview of notable treatment paradigms that have demonstrated considerable merit toward treating these disorders.
Collapse
Affiliation(s)
| | - Mariah S. Hahn
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, United States
| |
Collapse
|
819
|
Rahman Z, Bazaz MR, Devabattula G, Khan MA, Godugu C. Targeting H3K9 methyltransferase G9a and its related molecule GLP as a potential therapeutic strategy for cancer. J Biochem Mol Toxicol 2020; 35:e22674. [PMID: 33283949 DOI: 10.1002/jbt.22674] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 11/25/2020] [Indexed: 12/24/2022]
Abstract
H3K9 methyltransferase (G9a) and its relevant molecule GLP are the SET domain proteins that specifically add mono, di and trimethyl groups on to the histone H3K9, which lead to the transcriptional inactivation of chromatin and reduce the expression of cancer suppressor genes, which trigger growth and progress of several cancer types. Various studies have demonstrated that overexpression of H3K9 methyltransferase G9a and GLP in different kinds of tumors, like lung, breast, bladder, colon, cervical, gastric, skin cancers, hepatocellular carcinoma and hematological malignancies. Several G9a and GLP inhibitors such as BIX-01294, UNC0642, A-366 and DCG066 were developed to combat various cancers; however, there is a need for more effective and less toxic compounds. The current molecular docking study suggested that the selected new compounds such as ninhydrin, naphthoquinone, cysteamine and disulfide cysteamine could be suitable molecules as a G9a and GLP inhibitors. Furthermore, detailed cell based and preclinical animal studies are required to confirm their properties. In the current review, we discussed the role of G9a and GLP mediated epigenetic regulation in the cancers. A thorough literature review was done related to G9a and GLP. The databases used extensively for retrieval of information were PubMed, Medline, Scopus and Science-direct. Further, molecular docking was performed using Maestro Schrodinger version 9.2 software to investigate the binding profile of compounds with Human G9a HMT (PDB ID: 3FPD, 3RJW) and Human GLP MT (PDB ID: 6MBO, 6MBP).
Collapse
Affiliation(s)
- Ziaur Rahman
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Mohd Rabi Bazaz
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Geetanjali Devabattula
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Mohd Abrar Khan
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Chandraiah Godugu
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| |
Collapse
|
820
|
FAM122A is required for hematopoietic stem cell function. Leukemia 2020; 35:2130-2134. [PMID: 33262527 DOI: 10.1038/s41375-020-01099-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 10/14/2020] [Accepted: 11/15/2020] [Indexed: 11/08/2022]
|
821
|
Kim SJ, Kim EM, Yamamoto M, Park H, Shin H. Engineering Multi-Cellular Spheroids for Tissue Engineering and Regenerative Medicine. Adv Healthc Mater 2020; 9:e2000608. [PMID: 32734719 DOI: 10.1002/adhm.202000608] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/09/2020] [Indexed: 02/06/2023]
Abstract
Multi-cellular spheroids are formed as a 3D structure with dense cell-cell/cell-extracellular matrix interactions, and thus, have been widely utilized as implantable therapeutics and various ex vivo tissue models in tissue engineering. In principle, spheroid culture methods maximize cell-cell cohesion and induce spontaneous cellular assembly while minimizing cellular interactions with substrates by using physical forces such as gravitational or centrifugal forces, protein-repellant biomaterials, and micro-structured surfaces. In addition, biofunctional materials including magnetic nanoparticles, polymer microspheres, and nanofiber particles are combined with cells to harvest composite spheroids, to accelerate spheroid formation, to increase the mechanical properties and viability of spheroids, and to direct differentiation of stem cells into desirable cell types. Biocompatible hydrogels are developed to produce microgels for the fabrication of size-controlled spheroids with high efficiency. Recently, spheroids have been further engineered to fabricate structurally and functionally reliable in vitro artificial 3D tissues of the desired shape with enhanced specific biological functions. This paper reviews the overall characteristics of spheroids and general/advanced spheroid culture techniques. Significant roles of functional biomaterials in advanced spheroid engineering with emphasis on the use of spheroids in the reconstruction of artificial 3D tissue for tissue engineering are also thoroughly discussed.
Collapse
Affiliation(s)
- Se-Jeong Kim
- Department of Bioengineering, Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
- BK21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
| | - Eun Mi Kim
- Department of Bioengineering, Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
- BK21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
| | - Masaya Yamamoto
- Department of Materials Processing, Graduate School of Engineering, Tohoku University, 6-6-02 Aramaki-aza Aoba, Aoba-ku, Sendai, 980-8579, Japan
- Biomedical Engineering for Diagnosis and Treatment, Graduate School of Biomedical Engineering, Tohoku University, 6-6-02 Aramaki-aza Aoba, Aoba-ku, Sendai, 980-8579, Japan
| | - Hansoo Park
- School of Integrative Engineering, College of Engineering, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, 06974, Republic of Korea
| | - Heungsoo Shin
- Department of Bioengineering, Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
- BK21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
- Institute of Nano Science & Technology (INST), Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
| |
Collapse
|
822
|
Giduthuri AT, Theodossiou SK, Schiele NR, Srivastava SK. Dielectrophoresis as a tool for electrophysiological characterization of stem cells. BIOPHYSICS REVIEWS 2020; 1:011304. [PMID: 38505626 PMCID: PMC10903368 DOI: 10.1063/5.0025056] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 11/20/2020] [Indexed: 03/21/2024]
Abstract
Dielectrophoresis (DEP), a nonlinear electrokinetic technique caused by Maxwell-Wagner interfacial polarization of neutral particles in an electrolyte solution, is a powerful cell manipulation method used widely for various applications such as enrichment, trapping, and sorting of heterogeneous cell populations. While conventional cell characterization and sorting methods require tagging or labeling of cells, DEP has the potential to manipulate cells in a label-free way. Due to its unique ability to characterize and sort cells without the need of labeling, there is renewed interest in using DEP for stem cell research and regenerative medicine. Stem cells have the potential to differentiate into various lineages, but achieving homogeneous cell phenotypes from an initially heterogeneous cell population is a challenge. Using DEP to efficiently and affordably identify, sort, and enrich either undifferentiated or differentiated stem cell populations in a label-free way would advance their potential uses for applications in tissue engineering and regenerative medicine. This review summarizes recent, significant research findings regarding the electrophysiological characterization of stem cells, with a focus on cellular dielectric properties, i.e., permittivity and conductivity, and on studies that have obtained these measurements using techniques that preserve cell viability, such as crossover frequency. Potential applications for DEP in regenerative medicine are also discussed. Overall, DEP is a promising technique and, when used to characterize, sort, and enrich stem cells, will advance stem cell-based regenerative therapies.
Collapse
Affiliation(s)
- Anthony T. Giduthuri
- Department of Chemical & Biological Engineering, University of Idaho, Moscow, Idaho 83844, USA
| | - Sophia K. Theodossiou
- Department of Chemical & Biological Engineering, University of Idaho, Moscow, Idaho 83844, USA
| | - Nathan R. Schiele
- Department of Chemical & Biological Engineering, University of Idaho, Moscow, Idaho 83844, USA
| | - Soumya K. Srivastava
- Department of Chemical & Biological Engineering, University of Idaho, Moscow, Idaho 83844, USA
| |
Collapse
|
823
|
Embedding cells within nanoscale, rapidly mineralizing hydrogels: A new paradigm to engineer cell-laden bone-like tissue. J Struct Biol 2020; 212:107636. [PMID: 33039511 DOI: 10.1016/j.jsb.2020.107636] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 09/30/2020] [Accepted: 10/03/2020] [Indexed: 11/20/2022]
Abstract
Bone mineralization is a highly specific and dynamic nanoscale process that has been studied extensively from a structural, chemical, and biological standpoint. Bone tissue, therefore, may be defined by the interplay of its intricately mineralized matrix and the cells that regulate its biological function. However, the far majority of engineered bone model systems and bone replacement materials have been unable to replicate this key characteristic of bone tissue; that is, the ability of cells to be gradually and rapidly embedded in a three-dimensional (3D) heavily calcified matrix material. Here we review the characteristics that define the bone matrix from a nanostructural perspective. We then revisit the benefits and challenges of existing model systems and engineered bone replacement materials, and discuss recent efforts to replicate the biological, cellular, mechanical, and materials characteristics of bone tissue on the nano- to microscale. We pay particular attention to a recently proposed method developed by our group, which seeks to replicate key aspects of the entrapment of bone cells within a mineralized matrix with precisions down to the level of individual nano-crystallites, inclusive of the bone vasculature, and osteogenic differentiation process. In summary, this paper discusses existing and emerging evidence pointing towards future developments bridging the gap between the fields of biomineralization, structural biology, stem cells, and tissue engineering, which we believe will hold the key to engineer truly functional bone-like tissue in the laboratory.
Collapse
|
824
|
Abedi M, Alavi-Moghadam S, Payab M, Goodarzi P, Mohamadi-jahani F, Sayahpour FA, Larijani B, Arjmand B. Mesenchymal stem cell as a novel approach to systemic sclerosis; current status and future perspectives. CELL REGENERATION (LONDON, ENGLAND) 2020; 9:20. [PMID: 33258056 PMCID: PMC7704834 DOI: 10.1186/s13619-020-00058-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 09/03/2020] [Indexed: 12/13/2022]
Abstract
Systemic sclerosis is a rare chronic autoimmune disease with extensive microvascular injury, damage of endothelial cells, activation of immune responses, and progression of tissue fibrosis in the skin and various internal organs. According to epidemiological data, women's populations are more susceptible to systemic sclerosis than men. Until now, various therapeutic options are employed to manage the symptoms of the disease. Since stem cell-based treatments have developed as a novel approach to rescue from several autoimmune diseases, it seems that stem cells, especially mesenchymal stem cells as a powerful regenerative tool can also be advantageous for systemic sclerosis treatment via their remarkable properties including immunomodulatory and anti-fibrotic effects. Accordingly, we discuss the contemporary status and future perspectives of mesenchymal stem cell transplantation for systemic sclerosis.
Collapse
Affiliation(s)
- Mina Abedi
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Alavi-Moghadam
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Moloud Payab
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Parisa Goodarzi
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Fereshteh Mohamadi-jahani
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Forough Azam Sayahpour
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical sciences, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
825
|
Liang W, Chen X, Dong Y, Zhou P, Xu F. Recent advances in biomaterials as instructive scaffolds for stem cells in tissue repair and regeneration. INT J POLYM MATER PO 2020. [DOI: 10.1080/00914037.2020.1848832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Wenqing Liang
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, P. R. China
| | - Xuerong Chen
- Department of Orthopaedics, Shaoxing People’s Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing, P. R. China
| | - Yongqiang Dong
- Department of Orthopaedics, Xinchang People’s Hospital, Shaoxing, P. R. China
| | - Ping Zhou
- Department of Orthopaedics, Shaoxing People’s Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing, P. R. China
| | - Fangming Xu
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, P. R. China
| |
Collapse
|
826
|
Cox CR, Lynch S, Goldring C, Sharma P. Current Perspective: 3D Spheroid Models Utilizing Human-Based Cells for Investigating Metabolism-Dependent Drug-Induced Liver Injury. FRONTIERS IN MEDICAL TECHNOLOGY 2020; 2:611913. [PMID: 35047893 PMCID: PMC8757888 DOI: 10.3389/fmedt.2020.611913] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 11/04/2020] [Indexed: 12/21/2022] Open
Abstract
Drug-induced liver injury (DILI) remains a leading cause for the withdrawal of approved drugs. This has significant financial implications for pharmaceutical companies, places increasing strain on global health services, and causes harm to patients. For these reasons, it is essential that in-vitro liver models are capable of detecting DILI-positive compounds and their underlying mechanisms, prior to their approval and administration to patients or volunteers in clinical trials. Metabolism-dependent DILI is an important mechanism of drug-induced toxicity, which often involves the CYP450 family of enzymes, and is associated with the production of a chemically reactive metabolite and/or inefficient removal and accumulation of potentially toxic compounds. Unfortunately, many of the traditional in-vitro liver models fall short of their in-vivo counterparts, failing to recapitulate the mature hepatocyte phenotype, becoming metabolically incompetent, and lacking the longevity to investigate and detect metabolism-dependent DILI and those associated with chronic and repeat dosing regimens. Nevertheless, evidence is gathering to indicate that growing cells in 3D formats can increase the complexity of these models, promoting a more mature-hepatocyte phenotype and increasing their longevity, in vitro. This review will discuss the use of 3D in vitro models, namely spheroids, organoids, and perfusion-based systems to establish suitable liver models to investigate metabolism-dependent DILI.
Collapse
Affiliation(s)
- Christopher R. Cox
- Department of Pharmacology and Experimental Therapeutics, MRC Centre for Drug Safety Science, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, United Kingdom
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
- *Correspondence: Christopher R. Cox
| | - Stephen Lynch
- Department of Pharmacology and Experimental Therapeutics, MRC Centre for Drug Safety Science, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Christopher Goldring
- Department of Pharmacology and Experimental Therapeutics, MRC Centre for Drug Safety Science, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Parveen Sharma
- Department of Pharmacology and Experimental Therapeutics, MRC Centre for Drug Safety Science, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, United Kingdom
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
- Liverpool Centre for Cardiovascular Science, Liverpool, United Kingdom
| |
Collapse
|
827
|
Bashir NZ. The role of insulin-like growth factors in modulating the activity of dental mesenchymal stem cells. Arch Oral Biol 2020; 122:104993. [PMID: 33259987 DOI: 10.1016/j.archoralbio.2020.104993] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 11/14/2020] [Accepted: 11/19/2020] [Indexed: 12/27/2022]
Abstract
Regenerative treatment protocols are an exciting prospect in the management of oral pathology, as they allow for tissues to be restored to their original form and function, as compared to the reparative healing mechanisms which currently govern the outcomes of the majority of dental treatment. Stem cell therapy presents with a great deal of untapped potential in this pursuit of tissue regeneration, and, in particular, mesenchymal stem cells (MSCs) derived from dental tissues are of specific relevance with regards to their applications in engineering craniofacial tissues. A number of mediatory factors are involved in modulating the actions of dental MSCs, and, of these, insulin like growth factors (IGFs) are known to have potent effects in governing the behavior of these cells. The IGF family comprises a number of primary ligands, receptors, and binding proteins which are known to modulate the key properties of dental MSCs, such as their proliferation rates, differentiation potential, and mineralisation. The aims of this review are three-fold: (i) to present an overview of dental MSCs and the role of growth factors in modulating their characteristics, (ii) to discuss in greater detail the specific role of IGFs and the benefits they may convey for tissue engineering, and (iii) to provide a summary of potential for in vivo clinical translation of the current in vitro body of evidence.
Collapse
|
828
|
Samadani AA, Keymoradzdeh A, Shams S, Soleymanpour A, Rashidy-Pour A, Hashemian H, Vahidi S, Norollahi SE. CAR T-cells profiling in carcinogenesis and tumorigenesis: An overview of CAR T-cells cancer therapy. Int Immunopharmacol 2020; 90:107201. [PMID: 33249047 DOI: 10.1016/j.intimp.2020.107201] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/04/2020] [Accepted: 11/09/2020] [Indexed: 12/15/2022]
Abstract
Immunotherapy of cancer by chimeric antigen receptors (CAR) modified T-cell has a remarkable clinical potential for malignancies. Meaningly, it is a suitable cancer therapy to treat different solid tumors. CAR is a special recombinant protein combination with an antibody targeting structure alongside with signaling domain capacity on order to activate T cells. It is confirmed that the CAR-modified T cells have this ability to terminate and remove B cell malignancies. So, methodologies for investigations the pro risks and also strategies for neutralizing possible off-tumor consequences of are great importance successful protocols and strategies of CAR T-cell therapy can improve the efficacy and safety of this type of cancers. In this review article, we try to classify and illustrate main optimized plans in cancer CAR T-cell therapy.
Collapse
Affiliation(s)
- Ali Akbar Samadani
- Healthy Ageing Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran.
| | - Arman Keymoradzdeh
- Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Shima Shams
- Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Armin Soleymanpour
- Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Ali Rashidy-Pour
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
| | - Houman Hashemian
- Pediatrics Diseases Research Center, 17 Shahrivar Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Sogand Vahidi
- Clinical Research Development Unit of Poursina Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Seyedeh Elham Norollahi
- Clinical Research Development Unit of Poursina Hospital, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
829
|
Xie X, Chen J, Shu Z. From strict moral standards to ethical neutrality: a policy-guided shift in the patentability of human embryonic stem cells in China. Stem Cell Res Ther 2020; 11:499. [PMID: 33239089 PMCID: PMC7687789 DOI: 10.1186/s13287-020-02013-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 11/03/2020] [Indexed: 12/04/2022] Open
Abstract
Attitudes towards human embryonic stem cells (hESCs) in China have witnessed a significant shift in 2020 that can be attributed to China’s policy guidance. For ethical reasons, stricter standards are adopted to curb related regulations and patent licensing. Through the introduction of policies, some research on hESCs has been recognized as legitimate and feasible to a certain standard and scope. In the subsequent practice of patent examination, the dual influence of policy support and public interest has led to a shift in the examination standards of China’s intellectual property authority from “strict morality” to “ethical neutrality”, implying limited recognition of hESCs’ patentability. In view of the promotion of policy incentives for the transformation and application of corresponding research, there is considerable social demand to provide patent protection for research results. In this context, an adjustment of related regulations is illustrated in this revision, manifesting a partial shift in regulations towards a supportive stance consistent with policy.
Collapse
Affiliation(s)
- Xuekai Xie
- School of Law, East China University of Science and Technology, 130 Meilong Road, Xuhui District, Shanghai, China
| | - Jiajv Chen
- School of Law, Zhejiang University of Finance & Economics, Hangzhou, 18 Xueyuan Street, Jianggan District, Hangzhou, Zhejiang, China.
| | - Zhengyang Shu
- School of Law, Zhejiang University of Finance & Economics, Hangzhou, 18 Xueyuan Street, Jianggan District, Hangzhou, Zhejiang, China
| |
Collapse
|
830
|
Induced Pluripotency: A Powerful Tool for In Vitro Modeling. Int J Mol Sci 2020; 21:ijms21238910. [PMID: 33255453 PMCID: PMC7727808 DOI: 10.3390/ijms21238910] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 11/13/2020] [Accepted: 11/17/2020] [Indexed: 12/11/2022] Open
Abstract
One of the greatest breakthroughs of regenerative medicine in this century was the discovery of induced pluripotent stem cell (iPSC) technology in 2006 by Shinya Yamanaka. iPSCs originate from terminally differentiated somatic cells that have newly acquired the developmental capacity of self-renewal and differentiation into any cells of three germ layers. Before iPSCs can be used routinely in clinical practice, their efficacy and safety need to be rigorously tested; however, iPSCs have already become effective and fully-fledged tools for application under in vitro conditions. They are currently routinely used for disease modeling, preparation of difficult-to-access cell lines, monitoring of cellular mechanisms in micro- or macroscopic scales, drug testing and screening, genetic engineering, and many other applications. This review is a brief summary of the reprogramming process and subsequent differentiation and culture of reprogrammed cells into neural precursor cells (NPCs) in two-dimensional (2D) and three-dimensional (3D) conditions. NPCs can be used as biomedical models for neurodegenerative diseases (NDs), which are currently considered to be one of the major health problems in the human population.
Collapse
|
831
|
Matichescu A, Ardelean LC, Rusu LC, Craciun D, Bratu EA, Babucea M, Leretter M. Advanced Biomaterials and Techniques for Oral Tissue Engineering and Regeneration-A Review. MATERIALS (BASEL, SWITZERLAND) 2020; 13:E5303. [PMID: 33238625 PMCID: PMC7700200 DOI: 10.3390/ma13225303] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 11/15/2020] [Accepted: 11/19/2020] [Indexed: 12/11/2022]
Abstract
The reconstruction or repair of oral and maxillofacial functionalities and aesthetics is a priority for patients affected by tooth loss, congenital defects, trauma deformities, or various dental diseases. Therefore, in dental medicine, tissue reconstruction represents a major interest in oral and maxillofacial surgery, periodontics, orthodontics, endodontics, and even daily clinical practice. The current clinical approaches involve a vast array of techniques ranging from the traditional use of tissue grafts to the most innovative regenerative procedures, such as tissue engineering. In recent decades, a wide range of both artificial and natural biomaterials and scaffolds, genes, stem cells isolated from the mouth area (dental follicle, deciduous teeth, periodontal ligament, dental pulp, salivary glands, and adipose tissue), and various growth factors have been tested in tissue engineering approaches in dentistry, with many being proven successful. However, to fully eliminate the problems of traditional bone and tissue reconstruction in dentistry, continuous research is needed. Based on a recent literature review, this paper creates a picture of current innovative strategies applying dental stem cells for tissue regeneration in different dental fields and maxillofacial surgery, and offers detailed information regarding the available scientific data and practical applications.
Collapse
Affiliation(s)
- Anamaria Matichescu
- Department of Preventive Dentistry, Community and Oral Health, “Victor Babeș” University of Medicine and Pharmacy Timisoara, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania;
| | - Lavinia Cosmina Ardelean
- Department of Technology of Materials and Devices in Dental Medicine, “Victor Babeș” University of Medicine and Pharmacy Timisoara, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
| | - Laura-Cristina Rusu
- Department of Oral Pathology, “Victor Babeș” University of Medicine and Pharmacy Timisoara, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania; (L.-C.R.); (D.C.); (M.B.)
| | - Dragos Craciun
- Department of Oral Pathology, “Victor Babeș” University of Medicine and Pharmacy Timisoara, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania; (L.-C.R.); (D.C.); (M.B.)
| | - Emanuel Adrian Bratu
- Department of Implant Supported Restorations, “Victor Babeș” University of Medicine and Pharmacy Timisoara, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
| | - Marius Babucea
- Department of Oral Pathology, “Victor Babeș” University of Medicine and Pharmacy Timisoara, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania; (L.-C.R.); (D.C.); (M.B.)
| | - Marius Leretter
- Department of Prosthodontics, “Victor Babeș” University of Medicine and Pharmacy Timisoara, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania;
| |
Collapse
|
832
|
Ramakrishna RR, Abd Hamid Z, Wan Zaki WMD, Huddin AB, Mathialagan R. Stem cell imaging through convolutional neural networks: current issues and future directions in artificial intelligence technology. PeerJ 2020; 8:e10346. [PMID: 33240655 PMCID: PMC7680049 DOI: 10.7717/peerj.10346] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 10/21/2020] [Indexed: 12/12/2022] Open
Abstract
Stem cells are primitive and precursor cells with the potential to reproduce into diverse mature and functional cell types in the body throughout the developmental stages of life. Their remarkable potential has led to numerous medical discoveries and breakthroughs in science. As a result, stem cell-based therapy has emerged as a new subspecialty in medicine. One promising stem cell being investigated is the induced pluripotent stem cell (iPSC), which is obtained by genetically reprogramming mature cells to convert them into embryonic-like stem cells. These iPSCs are used to study the onset of disease, drug development, and medical therapies. However, functional studies on iPSCs involve the analysis of iPSC-derived colonies through manual identification, which is time-consuming, error-prone, and training-dependent. Thus, an automated instrument for the analysis of iPSC colonies is needed. Recently, artificial intelligence (AI) has emerged as a novel technology to tackle this challenge. In particular, deep learning, a subfield of AI, offers an automated platform for analyzing iPSC colonies and other colony-forming stem cells. Deep learning rectifies data features using a convolutional neural network (CNN), a type of multi-layered neural network that can play an innovative role in image recognition. CNNs are able to distinguish cells with high accuracy based on morphologic and textural changes. Therefore, CNNs have the potential to create a future field of deep learning tasks aimed at solving various challenges in stem cell studies. This review discusses the progress and future of CNNs in stem cell imaging for therapy and research.
Collapse
Affiliation(s)
- Ramanaesh Rao Ramakrishna
- Biomedical Science Programme and Centre for Diagnostic, Therapeutic and Investigative Science, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Zariyantey Abd Hamid
- Biomedical Science Programme and Centre for Diagnostic, Therapeutic and Investigative Science, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Wan Mimi Diyana Wan Zaki
- Department of Electrical, Electronic & Systems Engineering, Faculty of Engineering & Built Environment, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | - Aqilah Baseri Huddin
- Department of Electrical, Electronic & Systems Engineering, Faculty of Engineering & Built Environment, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | - Ramya Mathialagan
- Biomedical Science Programme and Centre for Diagnostic, Therapeutic and Investigative Science, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
833
|
Sugai H, Tomita S, Ishihara S, Yoshioka K, Kurita R. Microfluidic Sensing System with a Multichannel Surface Plasmon Resonance Chip: Damage-Free Characterization of Cells by Pattern Recognition. Anal Chem 2020; 92:14939-14946. [PMID: 33112611 DOI: 10.1021/acs.analchem.0c02220] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The development of a versatile sensing strategy for the damage-free characterization of cultured cells is of great importance for both fundamental biological research and industrial applications. Here, we present a pattern-recognition-based cell-sensing approach using a multichannel surface plasmon resonance (SPR) chip. The chip, in which five cysteine derivatives with different structures are immobilized on Au films, is capable of generating five unique SPR sensorgrams for the cell-secreted molecules that are contained in cell culture media. An automatic statistical program was built to acquire kinetic parameters from the SPR sensorgrams and to select optimal parameters as "pattern information" for subsequent multivariate analysis. Our system rapidly (∼10 min) provides the complex information by merely depositing a small amount of cell culture media (∼25 μL) onto the chip, and the amount of information obtained is comparable to that furnished by a combination of conventional laborious biochemical assays. This noninvasive pattern-recognition-based cell-sensing approach could potentially be employed as a versatile tool for characterizing cells.
Collapse
Affiliation(s)
- Hiroka Sugai
- Health and Medical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8566, Japan
| | - Shunsuke Tomita
- Health and Medical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8566, Japan.,DAILAB, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), National Institute of Advanced Industrial Science and Technology (AIST), Central 5-41, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan
| | - Sayaka Ishihara
- Health and Medical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8566, Japan
| | - Kyoko Yoshioka
- Health and Medical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8566, Japan
| | - Ryoji Kurita
- Health and Medical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8566, Japan.,DAILAB, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), National Institute of Advanced Industrial Science and Technology (AIST), Central 5-41, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan.,Faculty of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8573, Japan
| |
Collapse
|
834
|
Huang Z, Powell R, Phillips JB, Haastert-Talini K. Perspective on Schwann Cells Derived from Induced Pluripotent Stem Cells in Peripheral Nerve Tissue Engineering. Cells 2020; 9:E2497. [PMID: 33213068 PMCID: PMC7698557 DOI: 10.3390/cells9112497] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/13/2020] [Accepted: 11/16/2020] [Indexed: 02/06/2023] Open
Abstract
Schwann cells play a crucial role in successful peripheral nerve repair and regeneration by supporting both axonal growth and myelination. Schwann cells are therefore a feasible option for cell therapy treatment of peripheral nerve injury. However, sourcing human Schwann cells at quantities required for development beyond research is challenging. Due to their availability, rapid in vitro expansion, survival, and integration within the host tissue, stem cells have attracted considerable attention as candidate cell therapies. Among them, induced pluripotent stem cells (iPSCs) with the associated prospects for personalized treatment are a promising therapy to take the leap from bench to bedside. In this critical review, we firstly focus on the current knowledge of the Schwann cell phenotype in regard to peripheral nerve injury, including crosstalk with the immune system during peripheral nerve regeneration. Then, we review iPSC to Schwann cell derivation protocols and the results from recent in vitro and in vivo studies. We finally conclude with some prospects for the use of iPSCs in clinical settings.
Collapse
Affiliation(s)
- Zhong Huang
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, 30623 Hannover, Germany;
- Center for Systems Neuroscience (ZSN) Hannover, 30559 Hannover, Germany
| | - Rebecca Powell
- Department of Pharmacology, UCL School of Pharmacy, 29-39 Brunswick Square, London WC1N 1AX, UK;
- UCL Centre for Nerve Engineering, University College London, London WC1E 6BT, UK
| | - James B. Phillips
- Department of Pharmacology, UCL School of Pharmacy, 29-39 Brunswick Square, London WC1N 1AX, UK;
- UCL Centre for Nerve Engineering, University College London, London WC1E 6BT, UK
| | - Kirsten Haastert-Talini
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, 30623 Hannover, Germany;
- Center for Systems Neuroscience (ZSN) Hannover, 30559 Hannover, Germany
| |
Collapse
|
835
|
Abstract
A potential ability of stem cells (SCs) is to regenerate and repair tissues in the human body by providing great prospects for therapeutic applications in the field of medicine. Currently, SC therapy is used in various conditions like diabetes, neurodegenerative disorders, etc. but faces some limitations like patient biocompatibility and chances of cross-infection. SCs are further modulated with nanoconjugates to overcome such challenges and will offer an advantage in the treatment of COVID-19. This pandemic requires design and development of proper treatment to save the life of human beings. Advancements in SC-based nanoconjugated therapy will open new avenues and create a significant impact in the development of futuristic nanomedicine. It may also emerge as a potential therapy for the management of infection in patients suffering from SARS-CoV-2 and related diseases such as pneumonia and virus-induced lung injuries. Mechanisms of stem cell-based nanoconjugates for inhibition of replication of corona virus. ![]()
Collapse
|
836
|
Johnson IRD, Nguyen CT, Wise P, Grimm D. Implications of Altered Endosome and Lysosome Biology in Space Environments. Int J Mol Sci 2020; 21:ijms21218205. [PMID: 33147843 PMCID: PMC7663135 DOI: 10.3390/ijms21218205] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 10/30/2020] [Accepted: 10/31/2020] [Indexed: 12/17/2022] Open
Abstract
Space exploration poses multiple challenges for mankind, not only on a technical level but also to the entire physiology of the space traveller. The human system must adapt to several environmental stressors, microgravity being one of them. Lysosomes are ubiquitous to every cell and essential for their homeostasis, playing significant roles in the regulation of autophagy, immunity, and adaptation of the organism to changes in their environment, to name a few. Dysfunction of the lysosomal system leads to age-related diseases, for example bone loss, reduced immune response or cancer. As these conditions have been shown to be accelerated following exposure to microgravity, this review elucidates the lysosomal response to real and simulated microgravity. Microgravity activates the endo-lysosomal system, with resulting impacts on bone loss, muscle atrophy and stem cell differentiation. The investigation of lysosomal adaptation to microgravity can be beneficial in the search for new biomarkers or therapeutic approaches to several disease pathologies on earth as well as the potential to mitigate pathophysiology during spaceflight.
Collapse
Affiliation(s)
- Ian R. D. Johnson
- Research in Space Environments Group, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia;
- Correspondence:
| | - Catherine T. Nguyen
- Research in Space Environments Group, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia;
| | - Petra Wise
- Department of Hematology and Oncology, Children’s Hospital of Los Angeles, Los Angeles, CA 90027, USA;
| | - Daniela Grimm
- Department of Microgravity and Translational Regenerative Medicine, Clinic for Plastic, Aesthetic and Hand Surgery, Otto-von-Guericke-University Magdeburg, 39106 Magdeburg, Germany;
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| |
Collapse
|
837
|
Eckhart KE, Schmidt SJ, Starvaggi FA, Wolf ME, Vickery WM, Sydlik SA. Peptide- and Protein-Graphene Oxide Conjugate Materials for Controlling Mesenchymal Stem Cell Fate. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2020. [DOI: 10.1007/s40883-020-00182-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
838
|
Sabzevari R, Roushandeh AM, Mehdipour A, Alini M, Roudkenar MH. SA/G hydrogel containing hCAP-18/LL-37-engineered WJ-MSCs-derived conditioned medium promoted wound healing in rat model of excision injury. Life Sci 2020; 261:118381. [DOI: 10.1016/j.lfs.2020.118381] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/18/2020] [Accepted: 08/31/2020] [Indexed: 12/20/2022]
|
839
|
Kim SA, Park HY, Shin YW, Go EJ, Kim YJ, Kim YC, Shetty AA, Kim SJ. Hemovac blood after total knee arthroplasty as a source of stem cells. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1406. [PMID: 33313151 PMCID: PMC7723525 DOI: 10.21037/atm-20-2215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Background With increasing life expectancy, stem cell therapy is receiving increasing attention. However, its application is restricted by ethical concerns. Hence a need exists for design of safe procedures for stem cell procurement. Here, we investigated whether hemovac blood (HVB) is an appropriate stem cell source. Methods HVB concentrates (HVBCs) from 20 total knee arthroplasty (TKA) patients and bone marrow aspirate (BMA) concentrates (BMACs) from 15 patients who underwent knee cartilage repair were comparatively evaluated. A bone marrow aspiration needle was inserted into the anterior superior iliac spine. Aspiration was performed using a 50-mL syringe, including 4 mL of anticoagulant, followed by centrifugation to obtain BMACs. To obtain HVBCs, blood was aspirated from the hemovac immediately after TKA surgery. Different cell types were enumerated. Isolation of BMA and HVB mononuclear cells was performed using density gradient centrifugation. Non-hematopoietic fibroblast colonies were quantified by colony forming unit-fibroblast assay surface marker analysis of HVB, HVBC, BMA, and BMAC was performed via flow cytometry. Mesenchymal stem cells (MSCs) isolated from HVBCs and BMACs were examined for osteogenic, adipogenic, and chondrogenic differentiation potential. Gene expression analysis was performed by quantitative real-time polymerase chain reaction (qRT-PCR). Results The number of cells from HVB and HVBC was significantly lower than from BMA and BMAC; however, the number of colonies in HVBC and BMAC did not differ significantly (P>0.05). Isolated cells from both sources had a fibroblast-like appearance, adhered to culture flasks, and formed colonies. Under different culture conditions, MSC-specific surface markers (CD29, CD44, CD90, CD105), osteogenic markers [RUNX2, osteopontin, osteocalcin, and alkaline phosphatase (ALP)] and adipogenic markers (PPARγ and C/EBPα) were expressed. Moreover, SOX9, type II collagen, and aggrecan were significantly upregulated upon chondrogenic differentiation. Conclusions HVB from TKA patients is a useful source of stem cells for research.
Collapse
Affiliation(s)
- Seon Ae Kim
- Department of Orthopedic Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ho Youn Park
- Department of Orthopedic Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yong-Woon Shin
- Department of Orthopaedic Surgery, College of Medicine, The Inje University of Korea, Seoul, Republic of Korea
| | - Eun Jeong Go
- Department of Orthopedic Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Young Ju Kim
- Department of Nursing Education & Administration, Uijeongbu St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yoo Chang Kim
- Department of Orthopedic Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Asode Ananthram Shetty
- Canterbury Christ Church University, Faculty of Health and Wellbeing, Chatham Maritime, Kent, UK
| | - Seok Jung Kim
- Department of Orthopedic Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
840
|
Di Stefano AB, Grisafi F, Perez-Alea M, Castiglia M, Di Simone M, Meraviglia S, Cordova A, Moschella F, Toia F. Cell quality evaluation with gene expression analysis of spheroids (3D) and adherent (2D) adipose stem cells. Gene 2020; 768:145269. [PMID: 33148459 DOI: 10.1016/j.gene.2020.145269] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/14/2020] [Accepted: 10/20/2020] [Indexed: 01/16/2023]
Abstract
Adipose stem cells (ASCs) represent a reliable source of stem cells with a widely demonstrated potential in regenerative medicine and tissue engineering applications. New recent insights suggest that three-dimensional (3D) models may closely mimic the native tissue properties; spheroids from adipose derived stem cells (SASCs) exhibit enhanced regenerative abilities compared with those of 2D models. Stem cell therapy success is determined by "cell-quality"; for this reason, the involvement of stress signals and cellular aging need to be further investigated. Here, we performed a comparative analysis of genes connected with stemness, aging, telomeric length and oxidative stress, in 3D and 2D primary cultures. The expression levels of stemness-related markers and anti-aging Sirtuin1 were significantly up-regulated (P < 0.001) in SASCs-3D while gene expression of aging-related p16INK4a was increased in ASCs-2D (P < 0.001). The 3D and 2D cultures also had a different gene expression profile for genes related to telomere maintenance (Shelterin complex, RNA Binding proteins and DNA repair genes) (P < 0.01 and P < 0.001) and oxidative stress (aldehyde dehydrogenase class1 and 3) (P < 0.05, P < 0.01 and P < 0.001) and presented a striking large variation in their cellular redox state. Based on our findings, we propose a "cell quality" model of SASCs, highlighting a precise molecular expression of several genes involved with stemness (SOX2, POU5F1 and NANOG), anti-aging (SIRT1), oxidative stress (ALDH3) and telomeres maintenance.
Collapse
Affiliation(s)
- Anna Barbara Di Stefano
- BIOPLAST-Laboratory of BIOlogy and Regenerative Medicine-PLASTic Surgery, Plastic and Reconstructive Surgery, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy.
| | - Federica Grisafi
- BIOPLAST-Laboratory of BIOlogy and Regenerative Medicine-PLASTic Surgery, Plastic and Reconstructive Surgery, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Mileidys Perez-Alea
- Advanced BioDesign, Parc Technologique de Lyon, Woodstock - Bâtiment Cèdre 1, Saint Priest, France
| | - Marta Castiglia
- Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Marta Di Simone
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), University of Palermo, Palermo, Italy; Department of Biomedicine, Neurosciences and Advanced Diagnosis, University of Palermo, Palermo, Italy
| | - Serena Meraviglia
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), University of Palermo, Palermo, Italy; Department of Biomedicine, Neurosciences and Advanced Diagnosis, University of Palermo, Palermo, Italy
| | - Adriana Cordova
- BIOPLAST-Laboratory of BIOlogy and Regenerative Medicine-PLASTic Surgery, Plastic and Reconstructive Surgery, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy; Plastic and Reconstructive Surgery, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy; Plastic and Reconstructive Surgery, Department of Oncology, Azienda Ospedaliera Universitaria Policlinico "Paolo Giaccone", 90127 Palermo, Italy
| | - Francesco Moschella
- BIOPLAST-Laboratory of BIOlogy and Regenerative Medicine-PLASTic Surgery, Plastic and Reconstructive Surgery, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Francesca Toia
- BIOPLAST-Laboratory of BIOlogy and Regenerative Medicine-PLASTic Surgery, Plastic and Reconstructive Surgery, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy; Plastic and Reconstructive Surgery, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy; Plastic and Reconstructive Surgery, Department of Oncology, Azienda Ospedaliera Universitaria Policlinico "Paolo Giaccone", 90127 Palermo, Italy
| |
Collapse
|
841
|
Hu N, Yu T, Chen J, Zheng S, Yan H, Duan J. Oxycodone stimulates normal and malignant hematopoietic progenitors via opioid-receptor-independent-β-catenin activation. Biochem Biophys Res Commun 2020; 533:1457-1463. [PMID: 33268026 DOI: 10.1016/j.bbrc.2020.10.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 10/13/2020] [Indexed: 10/23/2022]
Abstract
Oxycodone is a common type of opioid used for the treatment of moderate to severe pain. Besides its analgesic effects on neuron cells, the effects of oxycodone on other cell types are yet to be elucidated. We previously demonstrated that oxycodone displayed both pro- and anti-cancer effects on bulk cancer cells. This work further investigated the effects of oxycodone on normal and malignant hematopoietic stem cells. Using hematopoietic CD34+ cells isolated from normal bone marrow (NBM) or patients with acute myeloid leukemia (AML), we showed that oxycodone activates hematopoietic cells regardless of cell development stage and malignant status. Oxycodone dose-dependently increases colony formation and self-renewal capacity of NBM and AML stem/progenitor cells, and promotes proliferation of AML bulk cells. NBM stem/progenitor cells are more sensitive to oxycodone than AML counterparts. In addition, oxycodone alleviates chemotherapy drug-induced toxicity in AML stem/progenitor cells. Mechanism studies demonstrate that oxycodone acts on hematopoietic cells in an opioid-receptor-independent manner. Oxycodone did not affect epithelial growth factor receptor (EGFR) signaling neither but stimulated Wnt/β-catenin signaling. Rescue studies via depleting β-catenin using genetic and pharmacological approaches confirmed that β-catenin was required for the activation of hematopoietic cells induced by oxycodone. Our work demonstrates 1) the protective role of oxycodone in malignant hematopoietic cells from chemotherapy; 2) stimulatory effects of oxycodone in normal hematopoietic stem cells; and 3) ability of oxycodone in Wnt signaling activation.
Collapse
Affiliation(s)
- Nianchun Hu
- Department of Anesthesiology, Wuhan Central Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Ting Yu
- Department of Anesthesiology, Wuhan Central Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Jingli Chen
- Department of Anesthesiology, Wuhan Central Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Shirong Zheng
- Department of Paediatrics, The Second Staff Hospital of Wuhan Iron and Steel Group Corporation, Wuhan, Hubei Province, China
| | - Hong Yan
- Department of Anesthesiology, Wuhan Central Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Ji'an Duan
- Department of Anesthesiology, Wuhan Central Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China.
| |
Collapse
|
842
|
Generation of Insulin-Producing Cells from Canine Adipose Tissue-Derived Mesenchymal Stem Cells. Stem Cells Int 2020; 2020:8841865. [PMID: 33133196 PMCID: PMC7591982 DOI: 10.1155/2020/8841865] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 09/30/2020] [Accepted: 10/03/2020] [Indexed: 12/18/2022] Open
Abstract
The potential of mesenchymal stem cells (MSCs) to differentiate into nonmesodermal cells such as pancreatic beta cells has been reported. New cell-based therapy using MSCs for diabetes mellitus is anticipated as an alternative treatment option to insulin injection or islet transplantation in both human and veterinary medicine. Several protocols were reported for differentiation of MSCs into insulin-producing cells (IPCs), but no studies have reported IPCs generated from canine MSCs. The purpose of this study was to generate IPCs from canine adipose tissue-derived MSCs (AT-MSCs) in vitro and to investigate the effects of IPC transplantation on diabetic mice in vivo. Culturing AT-MSCs with the differentiation protocol under a two-dimensional culture system did not produce IPCs. However, spheroid-like small clusters consisting of canine AT-MSCs and human recombinant peptide μ-pieces developed under a three-dimensional (3D) culture system were successfully differentiated into IPCs. The generated IPCs under 3D culture condition were stained with dithizone and anti-insulin antibody. Canine IPCs also showed gene expression typical for pancreatic beta cells and increased insulin secretion in response to glucose stimulation. The blood glucose levels in streptozotocin-induced diabetic mice were decreased after injection with the supernatant of canine IPCs, but the hyperglycemic states of diabetic mice were not improved after transplanting IPCs subcutaneously or intramesenterically. The histological examination showed that the transplanted small clusters of IPCs were successfully engrafted to the mice and included cells positive for insulin by immunofluorescence. Several factors, such as the transplanted cell number, the origin of AT-MSCs, and the differentiation protocol, were considered potential reasons for the inability to improve the hyperglycemic state after IPC transplantation. These findings suggest that canine AT-MSCs can be differentiated into IPCs under a 3D culture system and IPC transplantation may be a new treatment option for dogs with diabetes mellitus.
Collapse
|
843
|
Singh P, Singh A, Shah S, Vataliya J, Mittal A, Chitkara D. RNA Interference Nanotherapeutics for Treatment of Glioblastoma Multiforme. Mol Pharm 2020; 17:4040-4066. [PMID: 32902291 DOI: 10.1021/acs.molpharmaceut.0c00709] [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] [Indexed: 02/06/2023]
Abstract
Nucleic acid therapeutics for RNA interference (RNAi) are gaining attention in the treatment and management of several kinds of the so-called "undruggable" tumors via targeting specific molecular pathways or oncogenes. Synthetic ribonucleic acid (RNAs) oligonucleotides like siRNA, miRNA, shRNA, and lncRNA have shown potential as novel therapeutics. However, the delivery of such oligonucleotides is significantly hampered by their physiochemical (such as hydrophilicity, negative charge, and instability) and biopharmaceutical features (in vivo serum stability, fast renal clearance, interaction with extracellular proteins, and hindrance in cellular internalization) that markedly reduce their biological activity. Recently, several nanocarriers have evolved as suitable non-viral vectors for oligonucleotide delivery, which are known to either complex or conjugate with these oligonucleotides efficiently and also overcome the extracellular and intracellular barriers, thereby allowing access to the tumoral micro-environment for the better and desired outcome in glioblastoma multiforme (GBM). This Review focuses on the up-to-date advancements in the field of RNAi nanotherapeutics utilized for GBM treatment.
Collapse
Affiliation(s)
- Prabhjeet Singh
- Department of Pharmacy, Birla Institute of Technology and Science (BITS) Pilani, Pilani Campus, Vidya Vihar, Pilani - 333 031, Rajasthan, India
| | - Aditi Singh
- Department of Pharmacy, Birla Institute of Technology and Science (BITS) Pilani, Pilani Campus, Vidya Vihar, Pilani - 333 031, Rajasthan, India
| | - Shruti Shah
- Department of Pharmacy, Birla Institute of Technology and Science (BITS) Pilani, Pilani Campus, Vidya Vihar, Pilani - 333 031, Rajasthan, India
| | - Jalpa Vataliya
- Department of Pharmacy, Birla Institute of Technology and Science (BITS) Pilani, Pilani Campus, Vidya Vihar, Pilani - 333 031, Rajasthan, India
| | - Anupama Mittal
- Department of Pharmacy, Birla Institute of Technology and Science (BITS) Pilani, Pilani Campus, Vidya Vihar, Pilani - 333 031, Rajasthan, India
| | - Deepak Chitkara
- Department of Pharmacy, Birla Institute of Technology and Science (BITS) Pilani, Pilani Campus, Vidya Vihar, Pilani - 333 031, Rajasthan, India
| |
Collapse
|
844
|
Preconditioned and Genetically Modified Stem Cells for Myocardial Infarction Treatment. Int J Mol Sci 2020; 21:ijms21197301. [PMID: 33023264 PMCID: PMC7582407 DOI: 10.3390/ijms21197301] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 09/25/2020] [Accepted: 09/25/2020] [Indexed: 02/07/2023] Open
Abstract
Ischemic heart disease and myocardial infarction remain leading causes of mortality worldwide. Existing myocardial infarction treatments are incapable of fully repairing and regenerating the infarcted myocardium. Stem cell transplantation therapy has demonstrated promising results in improving heart function following myocardial infarction. However, poor cell survival and low engraftment at the harsh and hostile environment at the site of infarction limit the regeneration potential of stem cells. Preconditioning with various physical and chemical factors, as well as genetic modification and cellular reprogramming, are strategies that could potentially optimize stem cell transplantation therapy for clinical application. In this review, we discuss the most up-to-date findings related to utilizing preconditioned stem cells for myocardial infarction treatment, focusing mainly on preconditioning with hypoxia, growth factors, drugs, and biological agents. Furthermore, genetic manipulations on stem cells, such as the overexpression of specific proteins, regulation of microRNAs, and cellular reprogramming to improve their efficiency in myocardial infarction treatment, are discussed as well.
Collapse
|
845
|
Chota A, George BP, Abrahamse H. Potential Treatment of Breast and Lung Cancer Using Dicoma anomala, an African Medicinal Plant. Molecules 2020; 25:molecules25194435. [PMID: 32992537 PMCID: PMC7582250 DOI: 10.3390/molecules25194435] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/16/2020] [Accepted: 09/25/2020] [Indexed: 12/25/2022] Open
Abstract
Globally, cancer has been identified as one of the leading causes of death in public health. Its etiology is based on consistent exposure to carcinogenic. Plant-derived anticancer compounds are known to be less toxic to the normal cells and are classified into acetylenic compounds, phenolics, terpenes, and phytosterols. Dicoma anomala is a perennial herb belonging to the family Asteraceae and is widely distributed in Sub-Saharan Africa and used in the treatment of cancer, malaria, fever, diabetes, ulcers, cold, and cough. This review aimed at highlighting the benefits of D. anomala in various therapeutic applications with special reference to the treatment of cancers and the mechanisms through which the plant-derived agents induce cell death.
Collapse
Affiliation(s)
| | | | - Heidi Abrahamse
- Correspondence: ; Tel.: +27-11-559-6550; Fax: +27-11-559-6448
| |
Collapse
|
846
|
Granz CL, Gorji A. Dental stem cells: The role of biomaterials and scaffolds in developing novel therapeutic strategies. World J Stem Cells 2020; 12:897-921. [PMID: 33033554 PMCID: PMC7524692 DOI: 10.4252/wjsc.v12.i9.897] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/05/2020] [Accepted: 08/16/2020] [Indexed: 02/06/2023] Open
Abstract
Dental stem cells (DSCs) are self-renewable cells that can be obtained easily from dental tissues, and are a desirable source of autologous stem cells. The use of DSCs for stem cell transplantation therapeutic approaches is attractive due to their simple isolation, high plasticity, immunomodulatory properties, and multipotential abilities. Using appropriate scaffolds loaded with favorable biomolecules, such as growth factors, and cytokines, can improve the proliferation, differentiation, migration, and functional capacity of DSCs and can optimize the cellular morphology to build tissue constructs for specific purposes. An enormous variety of scaffolds have been used for tissue engineering with DSCs. Of these, the scaffolds that particularly mimic tissue-specific micromilieu and loaded with biomolecules favorably regulate angiogenesis, cell-matrix interactions, degradation of extracellular matrix, organized matrix formation, and the mineralization abilities of DSCs in both in vitro and in vivo conditions. DSCs represent a promising cell source for tissue engineering, especially for tooth, bone, and neural tissue restoration. The purpose of the present review is to summarize the current developments in the major scaffolding approaches as crucial guidelines for tissue engineering using DSCs and compare their effects in tissue and organ regeneration.
Collapse
Affiliation(s)
- Cornelia Larissa Granz
- Epilepsy Research Center, Westfälische Wilhelms-Universität Münster, Münster 48149, Germany
| | - Ali Gorji
- Epilepsy Research Center, Westfälische Wilhelms-Universität Münster, Münster 48149, Germany
| |
Collapse
|
847
|
Woods N, MacLoughlin R. Defining a Regulatory Strategy for ATMP/Aerosol Delivery Device Combinations in the Treatment of Respiratory Disease. Pharmaceutics 2020; 12:E922. [PMID: 32993197 PMCID: PMC7601063 DOI: 10.3390/pharmaceutics12100922] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 09/13/2020] [Accepted: 09/21/2020] [Indexed: 12/12/2022] Open
Abstract
Advanced Therapeutic Medicinal Products (ATMP) are a heterogenous group of investigational medicinal products at the forefront of innovative therapies with direct applicability in respiratory diseases. ATMPs include, but are not limited to, stem cells, their secretome, or extracellular vesicles, and each have shown some potential when delivered topically within the lung. This review focuses on that subset of ATMPs. One key mode of delivery that has enabling potential in ATMP validation is aerosol-mediated delivery. The selection of the most appropriate aerosol generator technology is influenced by several key factors, including formulation, patient type, patient intervention, and healthcare economics. The aerosol-mediated delivery of ATMPs has shown promise for the treatment of both chronic and acute respiratory disease in pre-clinical and clinical trials; however, in order for these ATMP device combinations to translate from the bench through to commercialization, they must meet the requirements set out by the various global regulatory bodies. In this review, we detail the potential for ATMP utility in the lungs and propose the nebulization of ATMPs as a viable route of administration in certain circumstances. Further, we provide insight to the current regulatory guidance for nascent ATMP device combination product development within the EU and US.
Collapse
Affiliation(s)
- Niamh Woods
- College of Medicine, Nursing & Health Sciences, National University of Ireland, H91 TK33 Galway, Ireland;
| | - Ronan MacLoughlin
- School of Pharmacy & Biomolecular Sciences, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland
- School of Pharmacy and Pharmaceutical Sciences, Trinity College, D02 PN40 Dublin, Ireland
- Aerogen Ltd., Galway Business Park, H91 HE94 Galway, Ireland
| |
Collapse
|
848
|
Lu M, Guo J, Wu B, Zhou Y, Wu M, Farzaneh M, Khoshnam SE. Mesenchymal Stem Cell-Mediated Mitochondrial Transfer: a Therapeutic Approach for Ischemic Stroke. Transl Stroke Res 2020; 12:212-229. [PMID: 32975692 DOI: 10.1007/s12975-020-00853-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 09/14/2020] [Accepted: 09/16/2020] [Indexed: 12/17/2022]
Abstract
Stroke is the leading cause of death and adult disability worldwide. Mitochondrial dysfunction is one of the hallmarks of stroke-induced neuronal death, and maintaining mitochondrial function is essential in cell survival and neurological progress following ischemic stroke. Stem cell-mediated mitochondrial transfer represents an emerging therapeutic approach for ischemic stroke. Accumulating evidence suggests that mesenchymal stem cells (MSCs) can directly transfer healthy mitochondria to damaged cells, and rescue mitochondrial damage-provoked tissue degeneration. This review summarizes the research on MSCs-mediated mitochondrial transfer as a therapeutic strategy against ischemic stroke.
Collapse
Affiliation(s)
- Meng Lu
- Academy of Traditional Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, China.,Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Shijiazhuang, 050091, China.,Department of Formulaology, Basic Medicine College, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Jindong Guo
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Shijiazhuang, 050091, China.,Department of Formulaology, Basic Medicine College, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Bowen Wu
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Shijiazhuang, 050091, China.,Department of Biochemistry, Basic Medicine College, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Yuhui Zhou
- Academy of Traditional Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, China.,Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Shijiazhuang, 050091, China.,Department of Formulaology, Basic Medicine College, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Mishan Wu
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Shijiazhuang, 050091, China. .,Department of Formulaology, Basic Medicine College, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China.
| | - Maryam Farzaneh
- Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyed Esmaeil Khoshnam
- Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
849
|
Lee HY, Hong IS. Targeting Liver Cancer Stem Cells: An Alternative Therapeutic Approach for Liver Cancer. Cancers (Basel) 2020; 12:cancers12102746. [PMID: 32987767 PMCID: PMC7598600 DOI: 10.3390/cancers12102746] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 09/18/2020] [Accepted: 09/22/2020] [Indexed: 12/12/2022] Open
Abstract
The first report of cancer stem cell (CSC) from Bruce et al. has demonstrated the relatively rare population of stem-like cells in acute myeloid leukemia (AML). The discovery of leukemic CSCs prompted further identification of CSCs in multiple types of solid tumor. Recently, extensive research has attempted to identity CSCs in multiple types of solid tumors in the brain, colon, head and neck, liver, and lung. Based on these studies, we hypothesize that the initiation and progression of most malignant tumors rely largely on the CSC population. Recent studies indicated that stem cell-related markers or signaling pathways, such as aldehyde dehydrogenase (ALDH), CD133, epithelial cell adhesion molecule (EpCAM), Wnt/β-catenin signaling, and Notch signaling, contribute to the initiation and progression of various liver cancer types. Importantly, CSCs are markedly resistant to conventional therapeutic approaches and current targeted therapeutics. Therefore, it is believed that selectively targeting specific markers and/or signaling pathways of hepatic CSCs is an effective therapeutic strategy for treating chemotherapy-resistant liver cancer. Here, we provide an overview of the current knowledge on the hepatic CSC hypothesis and discuss the specific surface markers and critical signaling pathways involved in the development and maintenance of hepatic CSC subpopulations.
Collapse
Affiliation(s)
- Hwa-Yong Lee
- Department of Biomedical Science, Jungwon University, 85 Goesan-eup, Munmu-ro, Goesan-gun, Chungcheongbuk-do 367700, Korea;
| | - In-Sun Hong
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Korea
- Department of Molecular Medicine, School of Medicine, Gachon University, Incheon 406840, Korea
- Correspondence: ; Tel.: +82-32-899-6315; Fax: +82-32-899-6350
| |
Collapse
|
850
|
Chen T, Ali Al-Radhawi M, Sontag ED. A mathematical model exhibiting the effect of DNA methylation on the stability boundary in cell-fate networks. Epigenetics 2020; 16:436-457. [PMID: 32842865 DOI: 10.1080/15592294.2020.1805686] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Cell-fate networks are traditionally studied within the framework of gene regulatory networks. This paradigm considers only interactions of genes through expressed transcription factors and does not incorporate chromatin modification processes. This paper introduces a mathematical model that seamlessly combines gene regulatory networks and DNA methylation (DNAm), with the goal of quantitatively characterizing the contribution of epigenetic regulation to gene silencing. The 'Basin of Attraction percentage' is introduced as a metric to quantify gene silencing abilities. As a case study, a computational and theoretical analysis is carried out for a model of the pluripotent stem cell circuit as well as a simplified self-activating gene model. The results confirm that the methodology quantitatively captures the key role that DNAm plays in enhancing the stability of the silenced gene state.
Collapse
Affiliation(s)
- Tianchi Chen
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - M Ali Al-Radhawi
- Department of Electrical and Computer Engineering, Northeastern University, Boston, MA, USA
| | - Eduardo D Sontag
- Department of Bioengineering, Northeastern University, Boston, MA, USA.,Department of Electrical and Computer Engineering, Northeastern University, Boston, MA, USA.,Laboratory of Systems Pharmacology, Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA
| |
Collapse
|