1
|
Takeda Y, Dai P. Functional roles of pantothenic acid, riboflavin, thiamine, and choline in adipocyte browning in chemically induced human brown adipocytes. Sci Rep 2024; 14:18252. [PMID: 39107469 PMCID: PMC11303702 DOI: 10.1038/s41598-024-69364-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024] Open
Abstract
Brown fat is a therapeutic target for the treatment of obesity-associated metabolic diseases. However, nutritional intervention strategies for increasing the mass and activity of human brown adipocytes have not yet been established. To identify vitamins required for brown adipogenesis and adipocyte browning, chemical compound-induced brown adipocytes (ciBAs) were converted from human dermal fibroblasts under serum-free and vitamin-free conditions. Choline was found to be essential for adipogenesis. Additional treatment with pantothenic acid (PA) provided choline-induced immature adipocytes with browning properties and metabolic maturation, including uncoupling protein 1 (UCP1) expression, lipolysis, and mitochondrial respiration. However, treatment with high PA concentrations attenuated these effects along with decreased glycolysis. Transcriptome analysis showed that a low PA concentration activated metabolic genes, including the futile creatine cycle-related thermogenic genes, which was reversed by a high PA concentration. Riboflavin treatment suppressed thermogenic gene expression and increased lipolysis, implying a metabolic pathway different from that of PA. Thiamine treatment slightly activated thermogenic genes along with decreased glycolysis. In summary, our results suggest that specific B vitamins and choline are uniquely involved in the regulation of adipocyte browning via cellular energy metabolism in a concentration-dependent manner.
Collapse
Affiliation(s)
- Yukimasa Takeda
- Department of Cellular Regenerative Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-Cho, Kawaramachi-Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan.
| | - Ping Dai
- Department of Cellular Regenerative Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-Cho, Kawaramachi-Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan.
| |
Collapse
|
2
|
Didiasova M, Banning A, Tikkanen R. Development of precision therapies for rare inborn errors of metabolism: Functional investigations in cell culture models. J Inherit Metab Dis 2024; 47:509-516. [PMID: 37606592 DOI: 10.1002/jimd.12674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 08/18/2023] [Accepted: 08/18/2023] [Indexed: 08/23/2023]
Abstract
Due to the low number of patients, rare genetic diseases are a special challenge for the development of therapies, especially for diseases that result from numerous, patient-specific pathogenic variants. Precision medicine makes use of various kinds of molecular information about a specific variant, so that the possibilities for an effective therapy based on the molecular features of the variants can be elucidated. The attention to personalized precision therapies has increased among scientists and clinicians, since the "single drug for all patients" approach does not allow the classification of individuals in subgroups according to the differences in the disease genotype or phenotype. This review article summarizes some approaches of personalized precision medicine that can be used for a cost-effective and fast development of therapies, even for single patients. We have focused on specific examples on inborn errors of metabolism, with special attention on drug repurposing. Furthermore, we provide an overview of cell culture models that are suitable for precision medicine approaches.
Collapse
Affiliation(s)
- Miroslava Didiasova
- Medical Faculty, Institute of Biochemistry, University of Giessen, Giessen, Germany
| | - Antje Banning
- Medical Faculty, Institute of Biochemistry, University of Giessen, Giessen, Germany
| | - Ritva Tikkanen
- Medical Faculty, Institute of Biochemistry, University of Giessen, Giessen, Germany
| |
Collapse
|
3
|
Haage V, Tuddenham JF, Comandante-Lou N, Bautista A, Monzel A, Chiu R, Fujita M, Garcia FG, Bhattarai P, Patel R, Buonfiglioli A, Idiarte J, Herman M, Rinderspacher A, Mela A, Zhao W, Argenziano MG, Furnari JL, Banu MA, Landry DW, Bruce JN, Canoll P, Zhang Y, Nuriel T, Kizil C, Sproul AA, de Witte LD, Sims PA, Menon V, Picard M, De Jager PL. A pharmacological toolkit for human microglia identifies Topoisomerase I inhibitors as immunomodulators for Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.06.579103. [PMID: 38370689 PMCID: PMC10871172 DOI: 10.1101/2024.02.06.579103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
While efforts to identify microglial subtypes have recently accelerated, the relation of transcriptomically defined states to function has been largely limited to in silico annotations. Here, we characterize a set of pharmacological compounds that have been proposed to polarize human microglia towards two distinct states - one enriched for AD and MS genes and another characterized by increased expression of antigen presentation genes. Using different model systems including HMC3 cells, iPSC-derived microglia and cerebral organoids, we characterize the effect of these compounds in mimicking human microglial subtypes in vitro. We show that the Topoisomerase I inhibitor Camptothecin induces a CD74high/MHChigh microglial subtype which is specialized in amyloid beta phagocytosis. Camptothecin suppressed amyloid toxicity and restored microglia back to their homeostatic state in a zebrafish amyloid model. Our work provides avenues to recapitulate human microglial subtypes in vitro, enabling functional characterization and providing a foundation for modulating human microglia in vivo.
Collapse
Affiliation(s)
- Verena Haage
- Center for Translational & Computational Neuroimmunology, Neuroimmunology Division, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, United States
| | - John F. Tuddenham
- Center for Translational & Computational Neuroimmunology, Neuroimmunology Division, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, United States
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, United States
| | - Natacha Comandante-Lou
- Center for Translational & Computational Neuroimmunology, Neuroimmunology Division, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, United States
| | - Alex Bautista
- Center for Translational & Computational Neuroimmunology, Neuroimmunology Division, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, United States
| | - Anna Monzel
- Department of Psychiatry, Division of Behavioral Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, USA
| | - Rebecca Chiu
- Center for Translational & Computational Neuroimmunology, Neuroimmunology Division, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, United States
| | - Masashi Fujita
- Center for Translational & Computational Neuroimmunology, Neuroimmunology Division, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, United States
| | - Frankie G. Garcia
- Center for Translational & Computational Neuroimmunology, Neuroimmunology Division, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, United States
| | - Prabesh Bhattarai
- Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, United States
| | - Ronak Patel
- Department of Pathology and Cell Biology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, United States
| | - Alice Buonfiglioli
- Department of Psychiatry, Icahn School of Medicine, 1460 Madison Avenue, New York, NY, 10029, United States
| | - Juan Idiarte
- Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, United States
| | - Mathieu Herman
- Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, United States
| | | | - Angeliki Mela
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Wenting Zhao
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, United States
| | - Michael G. Argenziano
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Julia L. Furnari
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Matei A. Banu
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Donald W. Landry
- Department of Medicine, Columbia University, New York, NY 10032, United States
| | - Jeffrey N. Bruce
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Peter Canoll
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Ya Zhang
- Center for Translational & Computational Neuroimmunology, Neuroimmunology Division, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, United States
| | - Tal Nuriel
- Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, United States
| | - Caghan Kizil
- Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, United States
| | - Andrew A. Sproul
- Department of Pathology and Cell Biology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, United States
| | - Lotje D. de Witte
- Department of Psychiatry, Icahn School of Medicine, 1460 Madison Avenue, New York, NY, 10029, United States
| | - Peter A. Sims
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, United States
| | - Vilas Menon
- Center for Translational & Computational Neuroimmunology, Neuroimmunology Division, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, United States
| | - Martin Picard
- Department of Psychiatry, Division of Behavioral Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, USA
- Department of Neurology, H. Houston Merritt Center, Columbia Translational Neuroscience Initiative, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, USA
- New York State Psychiatric Institute, New York, USA
- Robert N Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Philip L. De Jager
- Center for Translational & Computational Neuroimmunology, Neuroimmunology Division, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, United States
| |
Collapse
|
4
|
Arai K, Saito F, Miyazaki M, Kushige H, Izu Y, Maeta N, Yamazoe K. Small Molecules Temporarily Induce Neuronal Features in Adult Canine Dermal Fibroblasts. Int J Mol Sci 2023; 24:15804. [PMID: 37958789 PMCID: PMC10648228 DOI: 10.3390/ijms242115804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/28/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
Several methods have been developed to generate neurons from other cell types for performing regeneration therapy and in vitro studies of central nerve disease. Small molecules (SMs) can efficiently induce neuronal features in human and rodent fibroblasts without transgenes. Although canines have been used as a spontaneous disease model of human central nerve, efficient neuronal reprogramming method of canine cells have not been well established. We aimed to induce neuronal features in adult canine dermal fibroblasts (ACDFs) by SMs and assess the permanency of these changes. ACDFs treated with eight SMs developed a round-shaped cell body with branching processes and expressed neuronal proteins, including βIII-tubulin, microtubule-associated protein 2 (MAP2), and neurofilament-medium. Transcriptome profiling revealed the upregulation of neuron-related genes, such as SNAP25 and GRIA4, and downregulation of fibroblast-related genes, such as COL12A1 and CCN5. Calcium fluorescent imaging demonstrated an increase in intracellular Ca2+ concentration upon stimulation with glutamate and KCl. Although neuronal features were induced similarly in basement membrane extract droplet culture, they diminished after culturing without SMs or in vivo transplantation into an injured spinal cord. In conclusion, SMs temporarily induce neuronal features in ACDFs. However, the analysis of bottlenecks in the neuronal induction is crucial for optimizing the process.
Collapse
Affiliation(s)
- Kiyotaka Arai
- Department of Veterinary Surgery, Faculty of Veterinary Medicine, Okayama University of Science, 1-3 Ikoi-no-oka, Imabari 794-8555, Japan; (M.M.); (H.K.); (N.M.); (K.Y.)
| | - Fumiyo Saito
- Department of Toxicology, Faculty of Veterinary Medicine, Okayama University of Science, 1-3 Ikoi-no-oka, Imabari 794-8555, Japan;
| | - Masashi Miyazaki
- Department of Veterinary Surgery, Faculty of Veterinary Medicine, Okayama University of Science, 1-3 Ikoi-no-oka, Imabari 794-8555, Japan; (M.M.); (H.K.); (N.M.); (K.Y.)
| | - Haruto Kushige
- Department of Veterinary Surgery, Faculty of Veterinary Medicine, Okayama University of Science, 1-3 Ikoi-no-oka, Imabari 794-8555, Japan; (M.M.); (H.K.); (N.M.); (K.Y.)
| | - Yayoi Izu
- Department of Laboratory Animal Science, Faculty of Veterinary Medicine, Okayama University of Science, 1-3 Ikoi-no-oka, Imabari 794-8555, Japan;
| | - Noritaka Maeta
- Department of Veterinary Surgery, Faculty of Veterinary Medicine, Okayama University of Science, 1-3 Ikoi-no-oka, Imabari 794-8555, Japan; (M.M.); (H.K.); (N.M.); (K.Y.)
| | - Kazuaki Yamazoe
- Department of Veterinary Surgery, Faculty of Veterinary Medicine, Okayama University of Science, 1-3 Ikoi-no-oka, Imabari 794-8555, Japan; (M.M.); (H.K.); (N.M.); (K.Y.)
| |
Collapse
|
5
|
Álvarez-Córdoba M, Talaverón-Rey M, Povea-Cabello S, Cilleros-Holgado P, Gómez-Fernández D, Piñero-Pérez R, Reche-López D, Munuera-Cabeza M, Suárez-Carrillo A, Romero-González A, Romero-Domínguez JM, López-Cabrera A, Armengol JÁ, Sánchez-Alcázar JA. Patient-Derived Cellular Models for Polytarget Precision Medicine in Pantothenate Kinase-Associated Neurodegeneration. Pharmaceuticals (Basel) 2023; 16:1359. [PMID: 37895830 PMCID: PMC10609847 DOI: 10.3390/ph16101359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 09/21/2023] [Accepted: 09/21/2023] [Indexed: 10/29/2023] Open
Abstract
The term neurodegeneration with brain iron accumulation (NBIA) brings together a broad set of progressive and disabling neurological genetic disorders in which iron is deposited preferentially in certain areas of the brain. Among NBIA disorders, the most frequent subtype is pantothenate kinase-associated neurodegeneration (PKAN) caused by pathologic variants in the PANK2 gene codifying the enzyme pantothenate kinase 2 (PANK2). To date, there are no effective treatments to stop the progression of these diseases. This review discusses the utility of patient-derived cell models as a valuable tool for the identification of pharmacological or natural compounds for implementing polytarget precision medicine in PKAN. Recently, several studies have described that PKAN patient-derived fibroblasts present the main pathological features associated with the disease including intracellular iron overload. Interestingly, treatment of mutant cell cultures with various supplements such as pantothenate, pantethine, vitamin E, omega 3, α-lipoic acid L-carnitine or thiamine, improved all pathophysiological alterations in PKAN fibroblasts with residual expression of the PANK2 enzyme. The information provided by pharmacological screenings in patient-derived cellular models can help optimize therapeutic strategies in individual PKAN patients.
Collapse
Affiliation(s)
- Mónica Álvarez-Córdoba
- Andalusian Centre for Developmental Biology (CABD-CSIC-Pablo de Olavide University), 41013 Seville, Spain; (M.Á.-C.); (M.T.-R.); (S.P.-C.); (P.C.-H.); (D.G.-F.); (R.P.-P.); (D.R.-L.); (M.M.-C.); (A.S.-C.); (A.R.-G.); (J.M.R.-D.); (A.L.-C.)
| | - Marta Talaverón-Rey
- Andalusian Centre for Developmental Biology (CABD-CSIC-Pablo de Olavide University), 41013 Seville, Spain; (M.Á.-C.); (M.T.-R.); (S.P.-C.); (P.C.-H.); (D.G.-F.); (R.P.-P.); (D.R.-L.); (M.M.-C.); (A.S.-C.); (A.R.-G.); (J.M.R.-D.); (A.L.-C.)
| | - Suleva Povea-Cabello
- Andalusian Centre for Developmental Biology (CABD-CSIC-Pablo de Olavide University), 41013 Seville, Spain; (M.Á.-C.); (M.T.-R.); (S.P.-C.); (P.C.-H.); (D.G.-F.); (R.P.-P.); (D.R.-L.); (M.M.-C.); (A.S.-C.); (A.R.-G.); (J.M.R.-D.); (A.L.-C.)
| | - Paula Cilleros-Holgado
- Andalusian Centre for Developmental Biology (CABD-CSIC-Pablo de Olavide University), 41013 Seville, Spain; (M.Á.-C.); (M.T.-R.); (S.P.-C.); (P.C.-H.); (D.G.-F.); (R.P.-P.); (D.R.-L.); (M.M.-C.); (A.S.-C.); (A.R.-G.); (J.M.R.-D.); (A.L.-C.)
| | - David Gómez-Fernández
- Andalusian Centre for Developmental Biology (CABD-CSIC-Pablo de Olavide University), 41013 Seville, Spain; (M.Á.-C.); (M.T.-R.); (S.P.-C.); (P.C.-H.); (D.G.-F.); (R.P.-P.); (D.R.-L.); (M.M.-C.); (A.S.-C.); (A.R.-G.); (J.M.R.-D.); (A.L.-C.)
| | - Rocío Piñero-Pérez
- Andalusian Centre for Developmental Biology (CABD-CSIC-Pablo de Olavide University), 41013 Seville, Spain; (M.Á.-C.); (M.T.-R.); (S.P.-C.); (P.C.-H.); (D.G.-F.); (R.P.-P.); (D.R.-L.); (M.M.-C.); (A.S.-C.); (A.R.-G.); (J.M.R.-D.); (A.L.-C.)
| | - Diana Reche-López
- Andalusian Centre for Developmental Biology (CABD-CSIC-Pablo de Olavide University), 41013 Seville, Spain; (M.Á.-C.); (M.T.-R.); (S.P.-C.); (P.C.-H.); (D.G.-F.); (R.P.-P.); (D.R.-L.); (M.M.-C.); (A.S.-C.); (A.R.-G.); (J.M.R.-D.); (A.L.-C.)
| | - Manuel Munuera-Cabeza
- Andalusian Centre for Developmental Biology (CABD-CSIC-Pablo de Olavide University), 41013 Seville, Spain; (M.Á.-C.); (M.T.-R.); (S.P.-C.); (P.C.-H.); (D.G.-F.); (R.P.-P.); (D.R.-L.); (M.M.-C.); (A.S.-C.); (A.R.-G.); (J.M.R.-D.); (A.L.-C.)
| | - Alejandra Suárez-Carrillo
- Andalusian Centre for Developmental Biology (CABD-CSIC-Pablo de Olavide University), 41013 Seville, Spain; (M.Á.-C.); (M.T.-R.); (S.P.-C.); (P.C.-H.); (D.G.-F.); (R.P.-P.); (D.R.-L.); (M.M.-C.); (A.S.-C.); (A.R.-G.); (J.M.R.-D.); (A.L.-C.)
| | - Ana Romero-González
- Andalusian Centre for Developmental Biology (CABD-CSIC-Pablo de Olavide University), 41013 Seville, Spain; (M.Á.-C.); (M.T.-R.); (S.P.-C.); (P.C.-H.); (D.G.-F.); (R.P.-P.); (D.R.-L.); (M.M.-C.); (A.S.-C.); (A.R.-G.); (J.M.R.-D.); (A.L.-C.)
| | - Jose Manuel Romero-Domínguez
- Andalusian Centre for Developmental Biology (CABD-CSIC-Pablo de Olavide University), 41013 Seville, Spain; (M.Á.-C.); (M.T.-R.); (S.P.-C.); (P.C.-H.); (D.G.-F.); (R.P.-P.); (D.R.-L.); (M.M.-C.); (A.S.-C.); (A.R.-G.); (J.M.R.-D.); (A.L.-C.)
| | - Alejandra López-Cabrera
- Andalusian Centre for Developmental Biology (CABD-CSIC-Pablo de Olavide University), 41013 Seville, Spain; (M.Á.-C.); (M.T.-R.); (S.P.-C.); (P.C.-H.); (D.G.-F.); (R.P.-P.); (D.R.-L.); (M.M.-C.); (A.S.-C.); (A.R.-G.); (J.M.R.-D.); (A.L.-C.)
| | - José Ángel Armengol
- Department of Physiology, Anatomy and Cellular Biology, Pablo de Olavide University, 41013 Seville, Spain;
| | - José Antonio Sánchez-Alcázar
- Andalusian Centre for Developmental Biology (CABD-CSIC-Pablo de Olavide University), 41013 Seville, Spain; (M.Á.-C.); (M.T.-R.); (S.P.-C.); (P.C.-H.); (D.G.-F.); (R.P.-P.); (D.R.-L.); (M.M.-C.); (A.S.-C.); (A.R.-G.); (J.M.R.-D.); (A.L.-C.)
| |
Collapse
|
6
|
Kurahashi T, Nishime C, Nishinaka E, Komaki Y, Seki F, Urano K, Harada Y, Yoshikawa T, Dai P. Transplantation of Chemical Compound-Induced Cells from Human Fibroblasts Improves Locomotor Recovery in a Spinal Cord Injury Rat Model. Int J Mol Sci 2023; 24:13853. [PMID: 37762156 PMCID: PMC10530737 DOI: 10.3390/ijms241813853] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 09/05/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
The development of regenerative medicine using cell therapy is eagerly awaited for diseases such as spinal cord injury (SCI), for which there has been no radical cure. We previously reported the direct conversion of human fibroblasts into neuronal-like cells using only chemical compounds; however, it is unclear whether chemical compound-induced neuronal-like (CiN) cells are clinically functional. In this study, we partially modified the method of inducing CiN cells (termed immature CiN cells) and examined their therapeutic efficacy, in a rat model of SCI, to investigate whether immature CiN cells are promising for clinical applications. Motor function recovery, after SCI, was assessed using the Basso, Beattie, and Bresnahan (BBB) test, as well as the CatWalk analysis. We found that locomotor recovery, after SCI in the immature CiN cell-transplanted group, was partially improved compared to that in the control group. Consistent with these results, magnetic resonance imaging (MRI) and histopathological analyses revealed that nerve recovery or preservation improved in the immature CiN cell-transplanted group. Furthermore, transcriptome analysis revealed that immature CiN cells highly express hepatocyte growth factor (HGF), which has recently been shown to be a promising therapeutic agent against SCI. Our findings suggest that immature CiN cells may provide an alternative strategy for the regenerative therapy of SCI.
Collapse
Affiliation(s)
- Toshihiro Kurahashi
- Department of Cellular Regenerative Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan; (T.K.); (T.Y.)
| | - Chiyoko Nishime
- Central Institute for Experimental Animals (CIEA), 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan; (C.N.); (E.N.); (Y.K.); (F.S.); (K.U.)
| | - Eiko Nishinaka
- Central Institute for Experimental Animals (CIEA), 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan; (C.N.); (E.N.); (Y.K.); (F.S.); (K.U.)
| | - Yuji Komaki
- Central Institute for Experimental Animals (CIEA), 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan; (C.N.); (E.N.); (Y.K.); (F.S.); (K.U.)
| | - Fumiko Seki
- Central Institute for Experimental Animals (CIEA), 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan; (C.N.); (E.N.); (Y.K.); (F.S.); (K.U.)
| | - Koji Urano
- Central Institute for Experimental Animals (CIEA), 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan; (C.N.); (E.N.); (Y.K.); (F.S.); (K.U.)
| | - Yoshinori Harada
- Department of Pathology and Cell Regulation, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan;
| | - Toshikazu Yoshikawa
- Department of Cellular Regenerative Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan; (T.K.); (T.Y.)
- Louis Pasteur Center for Medical Research, 103-5 Tanaka-Monzen-cho, Sakyo-ku, Kyoto 606-8225, Japan
| | - Ping Dai
- Department of Cellular Regenerative Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan; (T.K.); (T.Y.)
| |
Collapse
|
7
|
Yang JH, Petty CA, Dixon-McDougall T, Lopez MV, Tyshkovskiy A, Maybury-Lewis S, Tian X, Ibrahim N, Chen Z, Griffin PT, Arnold M, Li J, Martinez OA, Behn A, Rogers-Hammond R, Angeli S, Gladyshev VN, Sinclair DA. Chemically induced reprogramming to reverse cellular aging. Aging (Albany NY) 2023; 15:5966-5989. [PMID: 37437248 PMCID: PMC10373966 DOI: 10.18632/aging.204896] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 07/04/2023] [Indexed: 07/14/2023]
Abstract
A hallmark of eukaryotic aging is a loss of epigenetic information, a process that can be reversed. We have previously shown that the ectopic induction of the Yamanaka factors OCT4, SOX2, and KLF4 (OSK) in mammals can restore youthful DNA methylation patterns, transcript profiles, and tissue function, without erasing cellular identity, a process that requires active DNA demethylation. To screen for molecules that reverse cellular aging and rejuvenate human cells without altering the genome, we developed high-throughput cell-based assays that distinguish young from old and senescent cells, including transcription-based aging clocks and a real-time nucleocytoplasmic compartmentalization (NCC) assay. We identify six chemical cocktails, which, in less than a week and without compromising cellular identity, restore a youthful genome-wide transcript profile and reverse transcriptomic age. Thus, rejuvenation by age reversal can be achieved, not only by genetic, but also chemical means.
Collapse
Affiliation(s)
- Jae-Hyun Yang
- Paul F. Glenn Center for Biology of Aging Research, Department of Genetics, Blavatnik Institute, Harvard Medical School (HMS), Boston, MA 02115, USA
| | - Christopher A. Petty
- Paul F. Glenn Center for Biology of Aging Research, Department of Genetics, Blavatnik Institute, Harvard Medical School (HMS), Boston, MA 02115, USA
| | - Thomas Dixon-McDougall
- Paul F. Glenn Center for Biology of Aging Research, Department of Genetics, Blavatnik Institute, Harvard Medical School (HMS), Boston, MA 02115, USA
| | - Maria Vina Lopez
- Molecular and Biomedical Sciences, University of Maine, Orono, ME 04467, USA
| | - Alexander Tyshkovskiy
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow 119234, Russia
| | - Sun Maybury-Lewis
- Paul F. Glenn Center for Biology of Aging Research, Department of Genetics, Blavatnik Institute, Harvard Medical School (HMS), Boston, MA 02115, USA
| | - Xiao Tian
- Paul F. Glenn Center for Biology of Aging Research, Department of Genetics, Blavatnik Institute, Harvard Medical School (HMS), Boston, MA 02115, USA
| | - Nabilah Ibrahim
- Paul F. Glenn Center for Biology of Aging Research, Department of Genetics, Blavatnik Institute, Harvard Medical School (HMS), Boston, MA 02115, USA
| | - Zhili Chen
- Paul F. Glenn Center for Biology of Aging Research, Department of Genetics, Blavatnik Institute, Harvard Medical School (HMS), Boston, MA 02115, USA
| | - Patrick T. Griffin
- Paul F. Glenn Center for Biology of Aging Research, Department of Genetics, Blavatnik Institute, Harvard Medical School (HMS), Boston, MA 02115, USA
| | - Matthew Arnold
- Paul F. Glenn Center for Biology of Aging Research, Department of Genetics, Blavatnik Institute, Harvard Medical School (HMS), Boston, MA 02115, USA
| | - Jien Li
- Paul F. Glenn Center for Biology of Aging Research, Department of Genetics, Blavatnik Institute, Harvard Medical School (HMS), Boston, MA 02115, USA
| | - Oswaldo A. Martinez
- Paul F. Glenn Center for Biology of Aging Research, Department of Genetics, Blavatnik Institute, Harvard Medical School (HMS), Boston, MA 02115, USA
- Department of Biology and Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Alexander Behn
- Paul F. Glenn Center for Biology of Aging Research, Department of Genetics, Blavatnik Institute, Harvard Medical School (HMS), Boston, MA 02115, USA
| | - Ryan Rogers-Hammond
- Paul F. Glenn Center for Biology of Aging Research, Department of Genetics, Blavatnik Institute, Harvard Medical School (HMS), Boston, MA 02115, USA
| | - Suzanne Angeli
- Molecular and Biomedical Sciences, University of Maine, Orono, ME 04467, USA
| | - Vadim N. Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - David A. Sinclair
- Paul F. Glenn Center for Biology of Aging Research, Department of Genetics, Blavatnik Institute, Harvard Medical School (HMS), Boston, MA 02115, USA
| |
Collapse
|
8
|
Sun L, Yang N, Chen B, Bei Y, Kang Z, Zhang C, Zhang N, Xu P, Yang W, Wei J, Ke J, Sun W, Li X, Shen P. A novel mesenchymal stem cell-based regimen for acute myeloid leukemia differentiation therapy. Acta Pharm Sin B 2023; 13:3027-3042. [PMID: 37521858 PMCID: PMC10372914 DOI: 10.1016/j.apsb.2023.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 03/05/2023] [Accepted: 03/14/2023] [Indexed: 08/01/2023] Open
Abstract
Currently the main treatment of acute myeloid leukemia (AML) is chemotherapy combining hematopoietic stem cell transplantation. However, the unbearable side effect of chemotherapy and the high risk of life-threatening infections and disease relapse following hematopoietic stem cell transplantation restrict its application in clinical practice. Thus, there is an urgent need to develop alternative therapeutic tactics with significant efficacy and attenuated adverse effects. Here, we revealed that umbilical cord-derived mesenchymal stem cells (UC-MSC) efficiently induced AML cell differentiation by shuttling the neutrophil elastase (NE)-packaged extracellular vesicles (EVs) into AML cells. Interestingly, the generation and release of NE-packaged EVs could be dramatically increased by vitamin D receptor (VDR) activation in UC-MSC. Chemical activation of VDR by using its agonist 1α,25-dihydroxyvitamin D3 efficiently enhanced the pro-differentiation capacity of UC-MSC and then alleviated malignant burden in AML mouse model. Based on these discoveries, to evade the risk of hypercalcemia, we synthetized and identified sw-22, a novel non-steroidal VDR agonist, which exerted a synergistic pro-differentiation function with UC-MSC on mitigating the progress of AML. Collectively, our findings provided a non-gene editing MSC-based therapeutic regimen to overcome the differentiation blockade in AML.
Collapse
Affiliation(s)
- Luchen Sun
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Nanfei Yang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325035, China
| | - Bing Chen
- Department of Hematology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210093, China
| | - Yuncheng Bei
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210008, China
| | - Zisheng Kang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing 210009, China
| | - Can Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing 210009, China
| | - Nan Zhang
- Centre of Micro/Nano Manufacturing Technology (MNMT-Dublin), School of Mechanical & Materials Engineering, University College Dublin, Dublin 4, Ireland
| | - Peipei Xu
- Department of Hematology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210093, China
| | - Wei Yang
- Department of Surgery, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jia Wei
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210008, China
| | - Jiangqiong Ke
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Weijian Sun
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Xiaokun Li
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health) & School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325035, China
| | - Pingping Shen
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, Nanjing 210008, China
- Shenzhen Research Institute of Nanjing University, Shenzhen 518000, China
| |
Collapse
|
9
|
Jeong D, Lee Y, Lee SW, Ham S, Lee M, Choi NY, Wu G, Scholer HR, Ko K. Homogeneity of XEN Cells Is Critical for Generation of Chemically Induced Pluripotent Stem Cells. Mol Cells 2023; 46:209-218. [PMID: 36852435 PMCID: PMC10086553 DOI: 10.14348/molcells.2023.2127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 10/22/2022] [Accepted: 10/23/2022] [Indexed: 03/01/2023] Open
Abstract
In induced pluripotent stem cells (iPSCs), pluripotency is induced artificially by introducing the transcription factors Oct4, Sox2, Klf4, and c-Myc. When a transgene is introduced using a viral vector, the transgene may be integrated into the host genome and cause a mutation and cancer. No integration occurs when an episomal vector is used, but this method has a limitation in that remnants of the virus or vector remain in the cell, which limits the use of such iPSCs in therapeutic applications. Chemical reprogramming, which relies on treatment with small-molecule compounds to induce pluripotency, can overcome this problem. In this method, reprogramming is induced according to the gene expression pattern of extra-embryonic endoderm (XEN) cells, which are used as an intermediate stage in pluripotency induction. Therefore, iPSCs can be induced only from established XEN cells. We induced XEN cells using small molecules that modulate a signaling pathway and affect epigenetic modifications, and devised a culture method in which can be produced homogeneous XEN cells. At least 4 passages were required to establish morphologically homogeneous chemically induced XEN (CiXEN) cells, whose properties were similar to those of XEN cells, as revealed through cellular and molecular characterization. Chemically iPSCs derived from CiXEN cells showed characteristics similar to those of mouse embryonic stem cells. Our results show that the homogeneity of CiXEN cells is critical for the efficient induction of pluripotency by chemicals.
Collapse
Affiliation(s)
- Dahee Jeong
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul 05029, Korea
- Center for Stem Cell Research, Institute of Advanced Biomedical Science, Konkuk University, Seoul 05029, Korea
| | - Yukyeong Lee
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul 05029, Korea
- Center for Stem Cell Research, Institute of Advanced Biomedical Science, Konkuk University, Seoul 05029, Korea
| | - Seung-Won Lee
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul 05029, Korea
- Center for Stem Cell Research, Institute of Advanced Biomedical Science, Konkuk University, Seoul 05029, Korea
| | - Seokbeom Ham
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul 05029, Korea
- Center for Stem Cell Research, Institute of Advanced Biomedical Science, Konkuk University, Seoul 05029, Korea
| | - Minseong Lee
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul 05029, Korea
- Center for Stem Cell Research, Institute of Advanced Biomedical Science, Konkuk University, Seoul 05029, Korea
| | - Na Young Choi
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul 05029, Korea
- Center for Stem Cell Research, Institute of Advanced Biomedical Science, Konkuk University, Seoul 05029, Korea
| | - Guangming Wu
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510320, China
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster 48149, Germany
| | - Hans R. Scholer
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster 48149, Germany
| | - Kinarm Ko
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul 05029, Korea
- Center for Stem Cell Research, Institute of Advanced Biomedical Science, Konkuk University, Seoul 05029, Korea
- Research Institute of Medical Science, Konkuk University, Seoul 05029, Korea
| |
Collapse
|
10
|
Liu X, Guo C, Leng T, Fan Z, Mai J, Chen J, Xu J, Li Q, Jiang B, Sai K, Yang W, Gu J, Wang J, Sun S, Chen Z, Zhong Y, Liang X, Chen C, Cai J, Lin Y, Liang J, Hu J, Yan G, Zhu W, Yin W. Differential regulation of H3K9/H3K14 acetylation by small molecules drives neuron-fate-induction of glioma cell. Cell Death Dis 2023; 14:142. [PMID: 36805688 PMCID: PMC9941105 DOI: 10.1038/s41419-023-05611-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 01/20/2023] [Accepted: 01/23/2023] [Indexed: 02/22/2023]
Abstract
Differentiation therapy using small molecules is a promising strategy for improving the prognosis of glioblastoma (GBM). Histone acetylation plays an important role in cell fate determination. Nevertheless, whether histone acetylation in specific sites determines GBM cells fate remains to be explored. Through screening from a 349 small molecule-library, we identified that histone deacetylase inhibitor (HDACi) MS-275 synergized with 8-CPT-cAMP was able to transdifferentiate U87MG GBM cells into neuron-like cells, which were characterized by cell cycle arrest, rich neuron biomarkers, and typical neuron electrophysiology. Intriguingly, acetylation tags of histone 3 at lysine 9 (H3K9ac) were decreased in the promoter of multiple oncogenes and cell cycle genes, while ones of H3K9ac and histone 3 at lysine 14 (H3K14ac) were increased in the promoter of neuron-specific genes. We then compiled a list of genes controlled by H3K9ac and H3K14ac, and proved that it is a good predictive power for pathologic grading and survival prediction. Moreover, cAMP agonist combined with HDACi also induced glioma stem cells (GSCs) to differentiate into neuron-like cells through the regulation of H3K9ac/K14ac, indicating that combined induction has the potential for recurrence-preventive application. Furthermore, the combination of cAMP activator plus HDACi significantly repressed the tumor growth in a subcutaneous GSC-derived tumor model, and temozolomide cooperated with the differentiation-inducing combination to prolong the survival in an orthotopic GSC-derived tumor model. These findings highlight epigenetic reprogramming through H3K9ac and H3K14ac as a novel approach for driving neuron-fate-induction of GBM cells.
Collapse
Affiliation(s)
- Xincheng Liu
- grid.12981.330000 0001 2360 039XDepartment of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080 P. R. China ,grid.284723.80000 0000 8877 7471Department of Emergency Medicine, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080 P. R. China
| | - Cui Guo
- grid.12981.330000 0001 2360 039XDepartment of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080 P. R. China
| | - Tiandong Leng
- grid.9001.80000 0001 2228 775XDepartment of Neuroscience, Morehouse School of Medicine, Atlanta, GA 30310 USA
| | - Zhen Fan
- grid.12981.330000 0001 2360 039XDepartment of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080 P. R. China
| | - Jialuo Mai
- grid.12981.330000 0001 2360 039XDepartment of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080 P. R. China
| | - Jiehong Chen
- grid.12981.330000 0001 2360 039XDepartment of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080 P. R. China
| | - Jinhai Xu
- grid.12981.330000 0001 2360 039XGuangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080 P. R. China
| | - Qianyi Li
- grid.12981.330000 0001 2360 039XGuangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080 P. R. China
| | - Bin Jiang
- grid.12981.330000 0001 2360 039XGuangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080 P. R. China
| | - Ke Sai
- grid.488530.20000 0004 1803 6191Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060 P. R. China ,grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060 P. R. China
| | - Wenzhuo Yang
- grid.488530.20000 0004 1803 6191Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060 P. R. China ,grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060 P. R. China
| | - Jiayu Gu
- grid.12981.330000 0001 2360 039XDepartment of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080 P. R. China
| | - Jingyi Wang
- grid.12981.330000 0001 2360 039XDepartment of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080 P. R. China
| | - Shuxin Sun
- grid.488530.20000 0004 1803 6191Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060 P. R. China ,grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060 P. R. China
| | - Zhijie Chen
- grid.488530.20000 0004 1803 6191Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060 P. R. China ,grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060 P. R. China
| | - Yingqian Zhong
- grid.12981.330000 0001 2360 039XDepartment of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080 P. R. China
| | - Xuanming Liang
- grid.12981.330000 0001 2360 039XDepartment of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080 P. R. China
| | - Chaoxin Chen
- grid.12981.330000 0001 2360 039XDepartment of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080 P. R. China
| | - Jing Cai
- grid.12981.330000 0001 2360 039XDepartment of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080 P. R. China
| | - Yuan Lin
- grid.12981.330000 0001 2360 039XDepartment of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080 P. R. China
| | - Jiankai Liang
- grid.12981.330000 0001 2360 039XDepartment of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080 P. R. China
| | - Jun Hu
- grid.12981.330000 0001 2360 039XDepartment of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080 P. R. China
| | - Guangmei Yan
- grid.12981.330000 0001 2360 039XDepartment of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080 P. R. China
| | - Wenbo Zhu
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, P. R. China.
| | - Wei Yin
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, P. R. China.
| |
Collapse
|
11
|
Takeda Y, Harada Y, Yoshikawa T, Dai P. Mitochondrial Energy Metabolism in the Regulation of Thermogenic Brown Fats and Human Metabolic Diseases. Int J Mol Sci 2023; 24:ijms24021352. [PMID: 36674862 PMCID: PMC9861294 DOI: 10.3390/ijms24021352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/06/2023] [Accepted: 01/09/2023] [Indexed: 01/13/2023] Open
Abstract
Brown fats specialize in thermogenesis by increasing the utilization of circulating blood glucose and fatty acids. Emerging evidence suggests that brown adipose tissue (BAT) prevents the incidence of obesity-associated metabolic diseases and several types of cancers in humans. Mitochondrial energy metabolism in brown/beige adipocytes regulates both uncoupling protein 1 (UCP1)-dependent and -independent thermogenesis for cold adaptation and the utilization of excess nutrients and energy. Many studies on the quantification of human BAT indicate that mass and activity are inversely correlated with the body mass index (BMI) and visceral adiposity. Repression is caused by obesity-associated positive and negative factors that control adipocyte browning, de novo adipogenesis, mitochondrial energy metabolism, UCP1 expression and activity, and noradrenergic response. Systemic and local factors whose levels vary between lean and obese conditions include growth factors, inflammatory cytokines, neurotransmitters, and metal ions such as selenium and iron. Modulation of obesity-associated repression in human brown fats is a promising strategy to counteract obesity and related metabolic diseases through the activation of thermogenic capacity. In this review, we highlight recent advances in mitochondrial metabolism, thermogenic regulation of brown fats, and human metabolic diseases.
Collapse
Affiliation(s)
- Yukimasa Takeda
- Department of Cellular Regenerative Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
- Correspondence: (Y.T.); (P.D.); Tel.: +81-75-251-5444 (Y.T.); +81-75-251-5135 (P.D.)
| | - Yoshinori Harada
- Department of Pathology and Cell Regulation, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Toshikazu Yoshikawa
- Department of Cellular Regenerative Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
- Louis Pasteur Center for Medical Research, 103-5 Tanaka-Monzen-cho, Sakyo-ku, Kyoto 606-8225, Japan
| | - Ping Dai
- Department of Cellular Regenerative Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
- Correspondence: (Y.T.); (P.D.); Tel.: +81-75-251-5444 (Y.T.); +81-75-251-5135 (P.D.)
| |
Collapse
|
12
|
Jin Y, Kim H, Min S, Choi YS, Seo SJ, Jeong E, Kim SK, Lee HA, Jo SH, Park JH, Park BW, Sim WS, Kim JJ, Ban K, Kim YG, Park HJ, Cho SW. Three-dimensional heart extracellular matrix enhances chemically induced direct cardiac reprogramming. SCIENCE ADVANCES 2022; 8:eabn5768. [PMID: 36516259 PMCID: PMC9750148 DOI: 10.1126/sciadv.abn5768] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 11/12/2022] [Indexed: 06/17/2023]
Abstract
Direct cardiac reprogramming has emerged as a promising therapeutic approach for cardiac regeneration. Full chemical reprogramming with small molecules to generate cardiomyocytes may be more amenable than genetic reprogramming for clinical applications as it avoids safety concerns associated with genetic manipulations. However, challenges remain regarding low conversion efficiency and incomplete cardiomyocyte maturation. Furthermore, the therapeutic potential of chemically induced cardiomyocytes (CiCMs) has not been investigated. Here, we report that a three-dimensional microenvironment reconstituted with decellularized heart extracellular matrix can enhance chemical reprogramming and cardiac maturation of fibroblasts to cardiomyocytes. The resultant CiCMs exhibit elevated cardiac marker expression, sarcomeric organization, and improved electrophysiological features and drug responses. We investigated the therapeutic potential of CiCMs reprogrammed in three-dimensional heart extracellular matrix in a rat model of myocardial infarction. Our platform can facilitate the use of CiCMs for regenerative medicine, disease modeling, and drug screening.
Collapse
Affiliation(s)
- Yoonhee Jin
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Hyeok Kim
- Department of Biomedicine and Health Sciences, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Sungjin Min
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Yi Sun Choi
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Seung Ju Seo
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Eunseon Jeong
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Su Kyeom Kim
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Hyang-Ae Lee
- Korea Institute of Toxicology, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Sung-Hyun Jo
- Department of Chemical Engineering, Soongsil University, Seoul 06978, Republic of Korea
| | - Jae-Hyun Park
- Department of Biomedicine and Health Sciences, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Bong-Woo Park
- Department of Biomedicine and Health Sciences, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Woo-Sup Sim
- Department of Biomedicine and Health Sciences, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jin-Ju Kim
- Department of Biomedicine and Health Sciences, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Kiwon Ban
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon 999077, Hong Kong
| | - Yun-Gon Kim
- Department of Chemical Engineering, Soongsil University, Seoul 06978, Republic of Korea
| | - Hun-Jun Park
- Department of Biomedicine and Health Sciences, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Cell Death Disease Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
13
|
Abbey D. Chemical journey of somatic cells to pluripotency. CELL REGENERATION 2022; 11:27. [PMID: 35918563 PMCID: PMC9346013 DOI: 10.1186/s13619-022-00126-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
AbstractReprogramming somatic cells to pluripotent stem cells has revolutionized the biomedical field by providing enormous hopes and opportunities for the regeneration of tissues and organs for transplantation. Using a small molecule cocktail of epigenetic modifiers and cell signalling inhibitors, a chemical-based easy and controllable technique for converting human somatic cells into chemically induced pluripotent stem cells was recently reported (Guan, Nature 605:325–31, 2022). This novel approach offers well-defined, safe, simple, easy, and clinical-grade manufacturing strategies for modifying the fate of human cells required for regenerative therapeutics.
Collapse
|
14
|
Wang Y, Wei T, Wang Q, Zhang C, Li K, Deng J. Resveratrol's neural protective effects for the injured embryoid body and cerebral organoid. BMC Pharmacol Toxicol 2022; 23:47. [PMID: 35820950 PMCID: PMC9275253 DOI: 10.1186/s40360-022-00593-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 07/06/2022] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE Resveratrol (RSV) is a polyphenol compound found in grapes, veratrum and other plants. It has been reported that RSV has anti-inflammatory, anti-oxidant, anti-cancer and other pharmacological effects. However, the impacts of RSV on development of nervous system are not understood well. The study aims to investigate RSV's neuroprotective effect during development and to provide a health care for pregnant women and their fetuses with RSV supplementation. METHODS In this study, we induced human induced pluripotent stem cells (hiPSCs) to form the embryoid bodies (EBs) and cerebral organoids (COs) with 3 dimensional (3D) culture. In the meantime, D-galactose (D-gal, 5 mg/ml) was used to make nervous injury model, and on the other hand, RSV with various doses, such as 2 μm/L, 10 μm/L, 50 μm/L, were applied to understand its neuroprotection. Therefore, the cultures were divided into control group, D-gal nervous injury group and RSV intervention groups. After that, the diameters of EBs and COs were measured regularly under a reverted microscope. In the meantime, the neural proliferation, cell apoptosis and the differentiation of germ layers were detected via immunofluorescence. RESULTS (1) D-gal could delay the development of EBs and COs; (2) RSV could rescue the atrophy of EBs and COs caused by D-gal; (3) RSV showed its neuroprotection, through promoting the neural cell proliferation, inhibiting apoptosis and accelerating the differentiation of germ layers. CONCLUSION RSV has a neuroprotective effect on the development of the nervous system, suggesting RSV supplementation may be necessary during the health care of pregnancy and childhood.
Collapse
Affiliation(s)
- Yanli Wang
- NHC Key Laboratory of Birth Defects Prevention, Henan Key Laboratory of Population Defects Prevention, Henan Institute of Reproduction Health Science and Technology, Zhengzhou, 450002, Henan Province, China
| | - Tingting Wei
- NHC Key Laboratory of Birth Defects Prevention, Henan Key Laboratory of Population Defects Prevention, Henan Institute of Reproduction Health Science and Technology, Zhengzhou, 450002, Henan Province, China
| | - Qiang Wang
- NHC Key Laboratory of Birth Defects Prevention, Henan Key Laboratory of Population Defects Prevention, Henan Institute of Reproduction Health Science and Technology, Zhengzhou, 450002, Henan Province, China
| | - Chaonan Zhang
- NHC Key Laboratory of Birth Defects Prevention, Henan Key Laboratory of Population Defects Prevention, Henan Institute of Reproduction Health Science and Technology, Zhengzhou, 450002, Henan Province, China
| | - Keyan Li
- NHC Key Laboratory of Birth Defects Prevention, Henan Key Laboratory of Population Defects Prevention, Henan Institute of Reproduction Health Science and Technology, Zhengzhou, 450002, Henan Province, China
| | - Jinbo Deng
- NHC Key Laboratory of Birth Defects Prevention, Henan Key Laboratory of Population Defects Prevention, Henan Institute of Reproduction Health Science and Technology, Zhengzhou, 450002, Henan Province, China.
| |
Collapse
|
15
|
Li ZH, Lu JD, Li SJ, Chen HL, Su ZJ. Generation of Leydig-like cells: approaches, characterization, and challenges. Asian J Androl 2022; 24:335-344. [PMID: 35017389 PMCID: PMC9295467 DOI: 10.4103/aja202193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 10/21/2021] [Indexed: 11/17/2022] Open
Abstract
Testosterone production by Leydig cells (LCs) plays a crucial role in male reproduction. The functional degeneration of LCs can cause testosterone deficiency, ultimately resulting in primary male hypogonadism. Transplantation of exogenous LCs with the ability to produce testosterone in response to the regulation of the hypothalamus-pituitary-gonad axis could be a promising alternative option to treat male primary hypogonadism. Recent studies have shown that it is possible to generate Leydig-like cells from stem cells by various approaches. In addition, somatic cells, such as embryonic or adult fibroblasts, have also been successfully reprogrammed into Leydig-like cells. In this review, we summarized the recent advances in the generation of Leydig-like cells, with an emphasis on comparing the effectiveness and safety of different protocols used and the cells generated. By further analyzing the characteristics of Leydig-like cells generated from fibroblasts based on small signaling molecules and regulatory factors, we found that although the cells may produce testosterone, they are significantly different from real LCs. For future in vivo applications, it is important that the steroidogenic cells generated be evaluated not only for their steroidogenic functions but also for their overall cell metabolic state by proteomics or transcriptomic tools.
Collapse
Affiliation(s)
- Zhao-Hui Li
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Department of Cell Biology, Jinan University, Guangzhou 510632, China
| | - Jun-Dong Lu
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Department of Cell Biology, Jinan University, Guangzhou 510632, China
| | - Shi-Jun Li
- Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China
| | - Hao-Lin Chen
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Zhi-Jian Su
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Department of Cell Biology, Jinan University, Guangzhou 510632, China
- National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou 510632, China
| |
Collapse
|
16
|
Wang X, Wu J, Wang W, Zhang Y, He D, Xiao B, Zhang H, Song A, Xing Y, Li B. Reprogramming of Rat Fibroblasts into Induced Neurons by Small-Molecule Compounds In Vitro and In Vivo. ACS Chem Neurosci 2022; 13:2099-2109. [PMID: 35723446 DOI: 10.1021/acschemneuro.2c00078] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Cell replacement is a promising approach for neurodegenerative disease treatment. Somatic cells such as fibroblasts can be induced to differentiate into neurons by specific transcription factors; however, the potential of viral vectors used for reprogramming to integrate into the genome raises concerns about the potential clinical applications of this approach. Here, we directly reprogrammed rat embryonic skin fibroblasts into induced neurons (iNs) via six small-molecule compounds (SMs) (VPA, CHIR99021, forskolin, Y-27632, Repsox, and P7C3-A20). iNs exhibit typical neuronal morphology, and immunofluorescence showed that more than 96% of the iNs expressed the early neuronal marker class III beta-tubulin (TUJ1) and that more than 91% of iNs expressed the mature neuronal marker neuron-specific enolase (NSE) after 10 days of reprogramming. Quantitative real-time polymerase chain reaction also showed that most iNs expressed the dopaminergic neuron marker tyrosine hydroxylase, the neural marker Nur correlation factor 1, the (γ-aminobutyric acid, GABA) GABAergic neuronal marker GABA, and the cholinergic neuron marker choline acetyltransferase. In addition, we found that cell proliferation decreased during reprogramming and that protein synthesis increased initially and then decreased. SMs were mixed with hydrogels, and the hydrogels were implanted subcutaneously into the backs of rats. After 7 days, the TUJ1 and NSE proteins were expressed in surrounding tissues, indicating that SMs caused reprogramming in vivo. In summary, rat skin fibroblasts can be efficiently reprogrammed into iNs by SMs in vitro and in vivo.
Collapse
Affiliation(s)
- Xueyun Wang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, P.R. China
| | - Jing Wu
- Department of Paediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001 Henan, P.R. China
| | - Wang Wang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, P.R. China
| | - Yuanwang Zhang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, P.R. China
| | - Dixin He
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, P.R. China
| | - Boying Xiao
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, P.R. China
| | - Haohao Zhang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, P.R. China
| | - Anqi Song
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, P.R. China
| | - Ying Xing
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, P.R. China
| | - Bo Li
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, P.R. China
| |
Collapse
|
17
|
Takeda Y, Dai P. Capsaicin directly promotes adipocyte browning in the chemical compound-induced brown adipocytes converted from human dermal fibroblasts. Sci Rep 2022; 12:6612. [PMID: 35459786 PMCID: PMC9033854 DOI: 10.1038/s41598-022-10644-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 04/12/2022] [Indexed: 01/01/2023] Open
Abstract
Human brown fat is a potential therapeutic target for preventing obesity and related metabolic diseases by dissipating energy as heat through uncoupling protein 1 (UCP1). We have previously reported a method to obtain chemical compound-induced brown adipocytes (ciBAs) converted from human dermal fibroblasts under serum-free conditions. However, pharmacological responses to bioactive molecules have been poorly characterised in ciBAs. This study showed that the treatment with Capsaicin, an agonist of transient receptor potential vanilloid 1, directly activated adipocyte browning such as UCP1 expression, mitochondrial biogenesis, energy consumption rates, and glycerol recycling in ciBAs. Furthermore, genome-wide transcriptome analysis indicated that Capsaicin activated a broad range of metabolic genes including glycerol kinase and glycerol 3-phosphate dehydrogenase 1, which could be associated with the activation of glycerol recycling and triglyceride synthesis. Capsaicin also activated UCP1 expression in immortalised human brown adipocytes but inhibited its expression in mesenchymal stem cell-derived adipocytes. Altogether, ciBAs successfully reflected the direct effects of Capsaicin on adipocyte browning. These findings suggested that ciBAs could serve as a promising cell model for screening of small molecules and dietary bioactive compounds targeting human brown adipocytes.
Collapse
Affiliation(s)
- Yukimasa Takeda
- Department of Cellular Regenerative Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan.
| | - Ping Dai
- Department of Cellular Regenerative Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan.
| |
Collapse
|
18
|
Generation of a Pure Culture of Neuron-like Cells with a Glutamatergic Phenotype from Mouse Astrocytes. Biomedicines 2022; 10:biomedicines10040928. [PMID: 35453678 PMCID: PMC9031297 DOI: 10.3390/biomedicines10040928] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 04/09/2022] [Accepted: 04/12/2022] [Indexed: 12/04/2022] Open
Abstract
Astrocyte-to-neuron reprogramming is a promising therapeutic approach for treatment of neurodegenerative diseases. The use of small molecules as an alternative to the virus-mediated ectopic expression of lineage-specific transcription factors negates the tumorigenic risk associated with viral genetic manipulation and uncontrolled differentiation of stem cells. However, because previously developed methods for small-molecule reprogramming of astrocytes to neurons are multistep, complex, and lengthy, their applications in biomedicine, including clinical treatment, are limited. Therefore, our objective in this study was to develop a novel chemical-based approach to the cellular reprogramming of astrocytes into neurons with high efficiency and low complexity. To accomplish that, we used C8-D1a, a mouse astrocyte cell line, to assess the role of small molecules in reprogramming protocols that otherwise suffer from inconsistencies caused by variations in donor of the primary cell. We developed a new protocol by which a chemical mixture formulated with Y26732, DAPT, RepSox, CHIR99021, ruxolitinib, and SAG rapidly and efficiently induced the neural reprogramming of astrocytes in four days, with a conversion efficiency of 82 ± 6%. Upon exposure to the maturation medium, those reprogrammed cells acquired a glutaminergic phenotype over the next eleven days. We also demonstrated the neuronal functionality of the induced cells by confirming KCL-induced calcium flux.
Collapse
|
19
|
Lin X, Rong C, Wu S. Two Sets of Compound Complex Driving for High Efficiency of Nonintegration Reprogramming of Human Fibroblasts. Cell Reprogram 2022; 24:71-79. [PMID: 35255219 DOI: 10.1089/cell.2021.0143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Currently, plentiful chemical-assisted methods have been applied for mouse induced pluripotent stem cells (iPSCs). It has been reported that small-molecule compounds can only reprogram mouse embryonic fibroblasts into mouse chemically induced pluripotent stem cells (mouse CiPSCs). However, human CiPSCs have not been reported. Therefore, it is still necessary to search for safer chemically assisted human pluripotent stem cells, which might realize the potential of human iPSCs. Here, we developed two sets of chemical cocktails to greatly improve the induction efficiency of human nonintegrated iPSCs, including the 4 compound mixture (4M) and the 5 compound mixture (4MI). These two sets of complex driving strategies might greatly improve the reprogramming efficiency to generate integration-free iPSCs.
Collapse
Affiliation(s)
- Xiangyi Lin
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,China-World Bright-Future Education Development Organization, Beijing, China
| | - Cuiping Rong
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shouhai Wu
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| |
Collapse
|
20
|
Zhong C, Liu M, Pan X, Zhu H. Tumorigenicity Risk of iPSCs in vivo: Nip it in the Bud. PRECISION CLINICAL MEDICINE 2022; 5:pbac004. [PMID: 35692443 PMCID: PMC9026204 DOI: 10.1093/pcmedi/pbac004] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/18/2022] [Accepted: 01/23/2022] [Indexed: 11/17/2022] Open
Abstract
In 2006, Takahashi and Yamanaka first created induced pluripotent stem cells from mouse fibroblasts via the retroviral introduction of genes encoding the transcription factors Oct3/4, Sox2, Klf44, and c-Myc. Since then, the future clinical application of somatic cell reprogramming technology has become an attractive research topic in the field of regenerative medicine. Of note, considerable interest has been placed in circumventing ethical issues linked to embryonic stem cell research. However, tumorigenicity, immunogenicity, and heterogeneity may hamper attempts to deploy this technology therapeutically. This review highlights the progress aimed at reducing induced pluripotent stem cells tumorigenicity risk and how to assess the safety of induced pluripotent stem cells cell therapy products.
Collapse
Affiliation(s)
- Chaoliang Zhong
- Department of Cell Biology, Naval Medical University, Shanghai, China
| | - Miao Liu
- Department of Cell Biology, Naval Medical University, Shanghai, China
| | - Xinghua Pan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, and Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong, China
- Shenzhen Bay Laboratory, Shenzhen 518032, Guangdong, China
| | - Haiying Zhu
- Department of Cell Biology, Naval Medical University, Shanghai, China
| |
Collapse
|
21
|
Chen G, Guo Y, Li C, Li S, Wan X. Small Molecules that Promote Self-Renewal of Stem Cells and Somatic Cell Reprogramming. Stem Cell Rev Rep 2021; 16:511-523. [PMID: 32185667 DOI: 10.1007/s12015-020-09965-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The ground state of embryonic stem cells (ESCs) is closely related to the development of regenerative medicine. Particularly, long-term culture of ESCs in vitro, maintenance of their undifferentiated state, self-renewal and multi-directional differentiation ability is the premise of ESCs mechanism and application research. Induced pluripotent stem cells (iPSC) reprogrammed from mouse embryonic fibroblasts (MEF) cells into cells with most of the ESC characteristics show promise towards solving ethical problems currently facing stem cell research. However, integration into chromosomal DNA through viral-mediated genes may activate proto oncogenes and lead to risk of cancer of iPSC. At the same time, iPS induction efficiency needs to be further improved to reduce the use of transcription factors. In this review, we discuss small molecules that promote self-renewal and reprogramming, including growth factor receptor inhibitors, GSK-3β and histone deacetylase inhibitors, metabolic regulators, pathway modulators as well as EMT/MET regulation inhibitors to enhance maintenance of ESCs and enable reprogramming. Additionally, we summarize the mechanism of action of small molecules on ESC self-renewal and iPSC reprogramming. Finally, we will report on the progress in identification of novel and potentially effective agents as well as selected strategies that show promise in regenerative medicine. On this basis, development of more small molecule combinations and efficient induction of chemically induced pluripotent stem cell (CiPSC) is vital for stem cell therapy. This will significantly improve research in pathogenesis, individualized drug screening, stem cell transplantation, tissue engineering and many other aspects.
Collapse
Affiliation(s)
- Guofang Chen
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China.
| | - Yu'e Guo
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Chao Li
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Shuangdi Li
- Departments of Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Xiaoping Wan
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China.
| |
Collapse
|
22
|
Singh B, Mal G, Verma V, Tiwari R, Khan MI, Mohapatra RK, Mitra S, Alyami SA, Emran TB, Dhama K, Moni MA. Stem cell therapies and benefaction of somatic cell nuclear transfer cloning in COVID-19 era. Stem Cell Res Ther 2021; 12:283. [PMID: 33980321 PMCID: PMC8114669 DOI: 10.1186/s13287-021-02334-5] [Citation(s) in RCA: 6] [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: 02/03/2021] [Accepted: 04/12/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The global health emergency of COVID-19 has necessitated the development of multiple therapeutic modalities including vaccinations, antivirals, anti-inflammatory, and cytoimmunotherapies, etc. COVID-19 patients suffer from damage to various organs and vascular structures, so they present multiple health crises. Mesenchymal stem cells (MSCs) are of interest to treat acute respiratory distress syndrome (ARDS) caused by SARS-CoV-2 infection. MAIN BODY Stem cell-based therapies have been verified for prospective benefits in copious preclinical and clinical studies. MSCs confer potential benefits to develop various cell types and organoids for studying virus-human interaction, drug testing, regenerative medicine, and immunomodulatory effects in COVID-19 patients. Apart from paving the ways to augment stem cell research and therapies, somatic cell nuclear transfer (SCNT) holds unique ability for a wide range of health applications such as patient-specific or isogenic cells for regenerative medicine and breeding transgenic animals for biomedical applications. Being a potent cell genome-reprogramming tool, the SCNT has increased prominence of recombinant therapeutics and cellular medicine in the current era of COVID-19. As SCNT is used to generate patient-specific stem cells, it avoids dependence on embryos to obtain stem cells. CONCLUSIONS The nuclear transfer cloning, being an ideal tool to generate cloned embryos, and the embryonic stem cells will boost drug testing and cellular medicine in COVID-19.
Collapse
Affiliation(s)
- Birbal Singh
- ICAR-Indian Veterinary Research Institute Regional Station, Palampur, Himachal Pradesh, India
| | - Gorakh Mal
- ICAR-Indian Veterinary Research Institute Regional Station, Palampur, Himachal Pradesh, India
| | - Vinod Verma
- Stem Cell Research Centre, Department of Hematology, Sanjay Gandhi Post-Graduate Institute of Medical Sciences, Lucknow, India
| | - Ruchi Tiwari
- Department of Veterinary Microbiology and Immunology, College of Veterinary Sciences, Uttar Pradesh Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go Anusandhan Sansthan (DUVASU), Mathura, 281001, India
| | - Muhammad Imran Khan
- Hefei National Lab for Physical Sciences at the Microscale and the Centers for Biomedical Engineering, University of Science and Technology of China, Hefei, China
| | - Ranjan K Mohapatra
- Department of Chemistry, Government College of Engineering, Keonjhar, Odisha, India
| | - Saikat Mitra
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Salem A Alyami
- Department of Mathematics and Statistics, Imam Mohammad Ibn Saud Islamic University, Riyadh, 11432, Saudi Arabia
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, 4381, Bangladesh.
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, 243 122, India.
| | - Mohammad Ali Moni
- WHO Collaborating Centre on eHealth, UNSW Digital Health, Faculty of Medicine, School of Public Health and Community Medicine, UNSW Sydney, Sydney, NSW, 2052, Australia.
| |
Collapse
|
23
|
Kostin A, Alam MA, McGinty D, Alam MN. Adult hypothalamic neurogenesis and sleep-wake dysfunction in aging. Sleep 2021; 44:5986548. [PMID: 33202015 DOI: 10.1093/sleep/zsaa173] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 07/22/2020] [Indexed: 12/21/2022] Open
Abstract
In the mammalian brain, adult neurogenesis has been extensively studied in the hippocampal sub-granular zone and the sub-ventricular zone of the anterolateral ventricles. However, growing evidence suggests that new cells are not only "born" constitutively in the adult hypothalamus, but many of these cells also differentiate into neurons and glia and serve specific functions. The preoptic-hypothalamic area plays a central role in the regulation of many critical functions, including sleep-wakefulness and circadian rhythms. While a role for adult hippocampal neurogenesis in regulating hippocampus-dependent functions, including cognition, has been extensively studied, adult hypothalamic neurogenic process and its contributions to various hypothalamic functions, including sleep-wake regulation are just beginning to unravel. This review is aimed at providing the current understanding of the hypothalamic adult neurogenic processes and the extent to which it affects hypothalamic functions, including sleep-wake regulation. We propose that hypothalamic neurogenic processes are vital for maintaining the proper functioning of the hypothalamic sleep-wake and circadian systems in the face of regulatory challenges. Sleep-wake disturbance is a frequent and challenging problem of aging and age-related neurodegenerative diseases. Aging is also associated with a decline in the neurogenic process. We discuss a hypothesis that a decrease in the hypothalamic neurogenic process underlies the aging of its sleep-wake and circadian systems and associated sleep-wake disturbance. We further discuss whether neuro-regenerative approaches, including pharmacological and non-pharmacological stimulation of endogenous neural stem and progenitor cells in hypothalamic neurogenic niches, can be used for mitigating sleep-wake and other hypothalamic dysfunctions in aging.
Collapse
Affiliation(s)
- Andrey Kostin
- Research Service (151A3), Veterans Affairs Greater Los Angeles Healthcare System, Sepulveda, CA
| | - Md Aftab Alam
- Research Service (151A3), Veterans Affairs Greater Los Angeles Healthcare System, Sepulveda, CA.,Department of Psychiatry, University of California, Los Angeles, CA
| | - Dennis McGinty
- Research Service (151A3), Veterans Affairs Greater Los Angeles Healthcare System, Sepulveda, CA.,Department of Psychology, University of California, Los Angeles, CA
| | - Md Noor Alam
- Research Service (151A3), Veterans Affairs Greater Los Angeles Healthcare System, Sepulveda, CA.,Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA
| |
Collapse
|
24
|
Min S, Lee HJ, Jin Y, Kim YH, Sung J, Choi HJ, Cho SW. Biphasic Electrical Pulse by a Micropillar Electrode Array Enhances Maturation and Drug Response of Reprogrammed Cardiac Spheroids. NANO LETTERS 2020; 20:6947-6956. [PMID: 32877191 DOI: 10.1021/acs.nanolett.0c01141] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Direct reprogramming is an efficient strategy to produce cardiac lineage cells necessary for cardiac tissue engineering and drug testing for cardiac toxicity. However, functional maturation of reprogrammed cardiomyocytes, which is of great importance for their regenerative potential and drug response, still remains challenging. In this study, we propose a novel electrode platform to promote direct cardiac reprogramming and improve the functionality of reprogrammed cardiac cells. Nonviral cardiac reprogramming was improved via a three-dimensional spheroid culture of chemically induced cardiomyocytes exposed to a small-molecule cocktail. A micropillar electrode array providing biphasic electrical pulses mimicking the heartbeat further enhanced maturation and electrophysiological properties of reprogrammed cardiac spheroids, leading to proper responses and increased sensitivity to drugs. On the basis of our results, we conclude that our device may have a wider application in the generation of functional cardiac cells for regenerative medicine and screening of novel drugs.
Collapse
Affiliation(s)
- Sungjin Min
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Hyo-Jung Lee
- Department of Materials Science and Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Yoonhee Jin
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Yu Heun Kim
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Jaesuk Sung
- Department of Materials Science and Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Heon-Jin Choi
- Department of Materials Science and Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
25
|
Kumar A, Mali P. Mapping regulators of cell fate determination: Approaches and challenges. APL Bioeng 2020; 4:031501. [PMID: 32637855 PMCID: PMC7332300 DOI: 10.1063/5.0004611] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 06/01/2020] [Indexed: 12/25/2022] Open
Abstract
Given the limited regenerative capacities of most organs, strategies are needed to efficiently generate large numbers of parenchymal cells capable of integration into the diseased organ. Although it was initially thought that terminally differentiated cells lacked the ability to transdifferentiate, it has since been shown that cellular reprogramming of stromal cells to parenchymal cells through direct lineage conversion holds great potential for the replacement of post-mitotic parenchymal cells lost to disease. To this end, an assortment of genetic, chemical, and mechanical cues have been identified to reprogram cells to different lineages both in vitro and in vivo. However, some key challenges persist that limit broader applications of reprogramming technologies. These include: (1) low reprogramming efficiencies; (2) incomplete functional maturation of derived cells; and (3) difficulty in determining the typically multi-factor combinatorial recipes required for successful transdifferentiation. To improve efficiency by comprehensively identifying factors that regulate cell fate, large scale genetic and chemical screening methods have thus been utilized. Here, we provide an overview of the underlying concept of cell reprogramming as well as the rationale, considerations, and limitations of high throughput screening methods. We next follow with a summary of unique hits that have been identified by high throughput screens to induce reprogramming to various parenchymal lineages. Finally, we discuss future directions of applying this technology toward human disease biology via disease modeling, drug screening, and regenerative medicine.
Collapse
Affiliation(s)
- Aditya Kumar
- Department of Bioengineering, University of California, San Diego, La Jolla, California 92093, USA
| | - Prashant Mali
- Department of Bioengineering, University of California, San Diego, La Jolla, California 92093, USA
| |
Collapse
|
26
|
Sallam A, Mousa SA. Neurodegenerative Diseases and Cell Reprogramming. Mol Neurobiol 2020; 57:4767-4777. [PMID: 32785825 DOI: 10.1007/s12035-020-02039-5] [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: 03/23/2020] [Accepted: 07/24/2020] [Indexed: 10/23/2022]
Abstract
Neurodegenerative diseases have different types according to the onset of the disease, the time course, and the underlying pathology. Although the dogma that brain cells cannot regenerate has changed, the normal regenerative process of the brain is usually not sufficient to restore brain tissue defects after different pathological insults. Stem cell therapy and more recently cell reprogramming could achieve success in the process of brain renewal. This review article presents recent advances of stem cell therapies in neurodegenerative diseases and the role of cell reprogramming in the scope of optimizing a confined condition that could direct signaling pathways of the cell toward a specific neural lineage. Further, we will discuss different types of transcriptional factors and their role in neural cell fate direction.
Collapse
Affiliation(s)
- Abeer Sallam
- Department of Physiology, Faculty of Medicine, Alexandria University, Governorate, Alexandria, Egypt.,Center of Excellence for Research in Regenerative Medicine and its Applications (CERRMA) Faculty of Medicine, Alexandria University, Alexandria, Governorate, Egypt
| | - Shaker A Mousa
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, 1 Discovery Drive, Rensselaer, NY, 12144, USA.
| |
Collapse
|
27
|
Sweat gland regeneration: Current strategies and future opportunities. Biomaterials 2020; 255:120201. [PMID: 32592872 DOI: 10.1016/j.biomaterials.2020.120201] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 04/21/2020] [Accepted: 06/09/2020] [Indexed: 12/13/2022]
Abstract
For patients with extensive skin defects, loss of sweat glands (SwGs) greatly decreases their quality of life. Indeed, difficulties in thermoregulation, ion reabsorption, and maintaining fluid balance might render them susceptible to hyperthermia, heatstroke, or even death. Despite extensive studies on the stem cell biology of the skin in recent years, in-situ regeneration of SwGs with both structural and functional fidelity is still challenging because of the limited regenerative capacity and cell fate control of resident progenitors. To overcome these challenges, one must consider both the intrinsic factors relevant to genetic and epigenetic regulation and cues from the cellular microenvironment. Here, we describe recent progress in molecular biology, developmental pathways, and cellular evolution associated with SwGdevelopment and maturation. This is followed by a summary of the current strategies used for cell-fate modulation, transmembrane drug delivery, and scaffold design associated with SwGregeneration. Finally, we offer perspectives for creating more sophisticated systems to accelerate patients' innate healing capacity and developing engineered skin constructs to treat or replace damaged tissues structurally and functionally.
Collapse
|
28
|
Liu K, Ma W, Li C, Li J, Zhang X, Liu J, Liu W, Wu Z, Zang C, Liang Y, Guo J, Li L. Advances in Transcription Factors Related to Neuroglial Cell Reprogramming. Transl Neurosci 2020; 11:17-27. [PMID: 32161682 PMCID: PMC7053399 DOI: 10.1515/tnsci-2020-0004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 01/07/2020] [Indexed: 11/27/2022] Open
Abstract
Neuroglial cells have a high level of plasticity, and many types of these cells are present in the nervous system. Neuroglial cells provide diverse therapeutic targets for neurological diseases and injury repair. Cell reprogramming technology provides an efficient pathway for cell transformation during neural regeneration, while transcription factor-mediated reprogramming can facilitate the understanding of how neuroglial cells mature into functional neurons and promote neurological function recovery.
Collapse
Affiliation(s)
- Kuangpin Liu
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan, China
| | - Wei Ma
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan, China
| | - Chunyan Li
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan, China
| | - Junjun Li
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan, China
| | - Xingkui Zhang
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan, China
| | - Jie Liu
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan, China
| | - Wei Liu
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan, China
| | - Zheng Wu
- Second Department of General Surgery, First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Chenghao Zang
- Second Department of General Surgery, First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Yu Liang
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan, China
| | - Jianhui Guo
- Second Department of General Surgery, First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Liyan Li
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
29
|
Yang Y, Wang QQ, Bozinov O, Xu RX, Sun YL, Wang SS. GSK‑3 inhibitor CHIR99021 enriches glioma stem‑like cells. Oncol Rep 2020; 43:1479-1490. [PMID: 32323804 PMCID: PMC7108089 DOI: 10.3892/or.2020.7525] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 02/14/2020] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma (GBM) is the most prevalent and lethal primary intrinsic brain cancer. The disease is essentially incurable, with glioblastomas characterized by resistance to both chemotherapy and radiotherapy, as well as by rapid tumor progression, all of which are mainly ascribed to glioma stem-like cells (GSLCs). In the present study, an improved model that is more similar to clinical GBM was constructed. Twenty clinical glioma samples were collected to obtain primary low-grade tumor cells. The cells were either maintained in serum-free medium as primary glioma-based cells (PGBCs) or cultured in the same medium with CHIR99021 as GSLCs. Then, the molecular and ultrastructural differences between the two cell groups were determined. Furthermore, the proliferation and migration of the GSLCs were examined and the potential mechanisms were investigated. Finally, temozolomide resistance in vitro and in the mouse model was assessed to study the properties of the induced GSLCs. The primary low-grade tumor cells extracted from surgical samples were enriched with GSLC properties, with high expression levels of CD133 and Nestin in 100 nM CHIR99021. The GSLCs exhibited high proliferation and migration. Furthermore, the expression of the PI3K/AKT signaling pathway and that of related genes and proteins were significantly enhanced by CHIR99021. The animal study also revealed high levels of STAT3, mTOR, NF-κB, and VEGF in the GSLC-transplanted mice. CHIR99021 could stably enhance GSLC properties in patient-derived glioma samples. It may provide a useful model for further study, helping to understand the pathogenesis of therapeutic resistance and to screen drug candidates.
Collapse
Affiliation(s)
- Yang Yang
- Department of Neurosurgery, University Hospital of Zurich, University of Zurich, CH‑8091 Zurich, Switzerland
| | - Qin-Qin Wang
- Neurosurgical Institute, The Seventh Medical Center of PLA Army General Hospital, Beijing 100700, P.R. China
| | - Oliver Bozinov
- Department of Neurosurgery, University Hospital of Zurich, University of Zurich, CH‑8091 Zurich, Switzerland
| | - Ru-Xiang Xu
- Neurosurgical Institute, The Seventh Medical Center of PLA Army General Hospital, Beijing 100700, P.R. China
| | - Yi-Lin Sun
- Neurosurgical Institute, The Seventh Medical Center of PLA Army General Hospital, Beijing 100700, P.R. China
| | - Shan-Shan Wang
- Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, P.R. China
| |
Collapse
|
30
|
A developed serum-free medium and an optimized chemical cocktail for direct conversion of human dermal fibroblasts into brown adipocytes. Sci Rep 2020; 10:3775. [PMID: 32111895 PMCID: PMC7048747 DOI: 10.1038/s41598-020-60769-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/17/2020] [Indexed: 01/12/2023] Open
Abstract
Brown adipocytes coordinate systemic energy metabolism associated with the pathogenesis of obesity and related metabolic diseases including type 2 diabetes. We have previously reported chemical compound-induced brown adipocytes (ciBAs) converted from human dermal fibroblasts without using transgenes. In this study, to reveal a precise molecular mechanism underlying the direct conversion and human adipocyte browning, we developed serum-free brown adipogenic medium (SFBAM) with an optimized chemical cocktail consisting of Rosiglitazone, Forskolin, and BMP7. During the direct conversion, treatment with BMP7 enhanced Ucp1 expression rather than the conversion efficiency in the absence of BMP signalling inhibitors. Moreover, treatment with a TGF-β signalling pathway inhibitor was no longer required in the serum-free medium, likely because the TGF-β pathway was already suppressed. SFBAM and the chemical cocktail efficiently converted human dermal fibroblasts into ciBAs within four weeks. The ciBAs exhibited increased mitochondrial levels, elevated oxygen consumption rate, and a response to β-adrenergic receptor agonists. Thus the ciBAs converted by the serum-free medium and the chemical cocktail provide a novel model of human brown (beige) adipocytes applicable for basic research, drug screening, and clinical applications.
Collapse
|
31
|
Ge JY, Zheng YW, Liu LP, Isoda H, Oda T. Impelling force and current challenges by chemicals in somatic cell reprogramming and expansion beyond hepatocytes. World J Stem Cells 2019; 11:650-665. [PMID: 31616541 PMCID: PMC6789182 DOI: 10.4252/wjsc.v11.i9.650] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 07/07/2019] [Accepted: 08/21/2019] [Indexed: 02/06/2023] Open
Abstract
In the field of regenerative medicine, generating numerous transplantable functional cells in the laboratory setting on a large scale is a major challenge. However, the in vitro maintenance and expansion of terminally differentiated cells are challenging because of the lack of specific environmental and intercellular signal stimulations, markedly hindering their therapeutic application. Remarkably, the generation of stem/progenitor cells or functional cells with effective proliferative potential is markedly in demand for disease modeling, cell-based transplantation, and drug discovery. Despite the potent genetic manipulation of transcription factors, integration-free chemically defined approaches for the conversion of somatic cell fate have garnered considerable attention in recent years. This review aims to summarize the progress thus far and discuss the advantages, limitations, and challenges of the impact of full chemicals on the stepwise reprogramming of pluripotency, direct lineage conversion, and direct lineage expansion on somatic cells. Owing to the current chemical-mediated induction, reprogrammed pluripotent stem cells with reproducibility difficulties, and direct lineage converted cells with marked functional deficiency, it is imperative to generate the desired cell types directly by chemically inducing their potent proliferation ability through a lineage-committed progenitor state, while upholding the maturation and engraftment capacity posttransplantation in vivo. Together with the comprehensive understanding of the mechanism of chemical drives, as well as the elucidation of specificity and commonalities, the precise manipulation of the expansion for diverse functional cell types could broaden the available cell sources and enhance the cellular function for clinical application in future.
Collapse
Affiliation(s)
- Jian-Yun Ge
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yun-Wen Zheng
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
- Institute of Regenerative Medicine and Affiliated Hospital, Jiangsu University, Zhenjiang 212001, Jiangsu Province, China
- Department of Regenerative Medicine, School of Medicine, Yokohama City University, Yokohama 236-0004, Japan
| | - Li-Ping Liu
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
- Institute of Regenerative Medicine and Affiliated Hospital, Jiangsu University, Zhenjiang 212001, Jiangsu Province, China
| | - Hiroko Isoda
- Faculty of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8572, Japan
| | - Tatsuya Oda
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
32
|
Gong L, Yan Q, Zhang Y, Fang X, Liu B, Guan X. Cancer cell reprogramming: a promising therapy converting malignancy to benignity. Cancer Commun (Lond) 2019; 39:48. [PMID: 31464654 PMCID: PMC6716904 DOI: 10.1186/s40880-019-0393-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 08/14/2019] [Indexed: 02/07/2023] Open
Abstract
In the past decade, remarkable progress has been made in reprogramming terminally differentiated somatic cells and cancer cells into induced pluripotent cells and cancer cells with benign phenotypes. Recent studies have explored various approaches to induce reprogramming from one cell type to another, including lineage-specific transcription factors-, combinatorial small molecules-, microRNAs- and embryonic microenvironment-derived exosome-mediated reprogramming. These reprogramming approaches have been proven to be technically feasible and versatile to enable re-activation of sequestered epigenetic regions, thus driving fate decisions of differentiated cells. One of the significant utilities of cancer cell reprogramming is the therapeutic potential of retrieving normal cell functions from various malignancies. However, there are several major obstacles to overcome in cancer cell reprogramming before clinical translation, including characterization of reprogramming mechanisms, improvement of reprogramming efficiency and safety, and development of delivery methods. Recently, several insights in reprogramming mechanism have been proposed, and determining progress has been achieved to promote reprogramming efficiency and feasibility, allowing it to emerge as a promising therapy against cancer in the near future. This review aims to discuss recent applications in cancer cell reprogramming, with a focus on the clinical significance and limitations of different reprogramming approaches, while summarizing vital roles played by transcription factors, small molecules, microRNAs and exosomes during the reprogramming process.
Collapse
Affiliation(s)
- Lanqi Gong
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, 999077, P.R. China.,State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, 999077, P.R. China
| | - Qian Yan
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, 999077, P.R. China.,State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, 999077, P.R. China
| | - Yu Zhang
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, 999077, P.R. China.,State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, 999077, P.R. China
| | - Xiaona Fang
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, 999077, P.R. China.,State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, 999077, P.R. China
| | - Beilei Liu
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, 999077, P.R. China.,State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, 999077, P.R. China
| | - Xinyuan Guan
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, 999077, P.R. China. .,State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, 999077, P.R. China.
| |
Collapse
|
33
|
Chang Y, Cho B, Kim S, Kim J. Direct conversion of fibroblasts to osteoblasts as a novel strategy for bone regeneration in elderly individuals. Exp Mol Med 2019; 51:1-8. [PMID: 31073120 PMCID: PMC6509166 DOI: 10.1038/s12276-019-0251-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 12/24/2018] [Accepted: 01/28/2019] [Indexed: 12/31/2022] Open
Abstract
Mortality caused by age-related bone fractures or osteoporosis is steadily increasing worldwide as the population ages. The pace of the development of bone regeneration engineering to treat bone fractures has consequently increased in recent years. A range of techniques for bone regeneration, such as immunotherapy, allografts, and hydrogel therapy, have been devised. Cell-based therapies using bone marrow-derived mesenchymal stem cells and induced pluripotent stem cells derived from somatic cells are considered to be suitable approaches for bone repair. However, these cell-based therapies suffer from a number of limitations in terms of efficiency and safety. Somatic cells can also be directly differentiated into osteoblasts by several transcription factors. As osteoblasts play a central role in the process of bone formation, the direct reprogramming of fibroblasts into osteoblasts may hence be a new way to treat bone fractures in elderly individuals. Here, we review recent developments regarding the therapeutic potential of the direct reprogramming of cells for bone regeneration. Reprogramming cells that produce connective tissue to form bone instead could help prevent fractures in the elderly. Bones weaken with age, and fractures are a significant health risk in ageing populations. Most current bone regeneration treatments use stem cells, which can differentiate into any type of cell and have infinite capacity to divide; however, they are difficult to source and can lead to tumor formation. Jongpil Kim at Dongguk University in South Korea and coworkers have reviewed a new method that uses genetic signals to transform connective tissue-forming cells into bone-producing cells. The reprogrammed cells have been shown to generate new bone at the desired site, and because they have already lost their capacity for infinite division, tumor formation risk is greatly reduced. This method shows promise to expand treatment options for fractures and osteoporosis.
Collapse
Affiliation(s)
- Yujung Chang
- Department of Biomedical Engineering, Dongguk University, Pildong-ro 1-gil 30, Jung-gu, Seoul, 04620, Republic of Korea
| | - Byounggook Cho
- Department of Biomedical Engineering, Dongguk University, Pildong-ro 1-gil 30, Jung-gu, Seoul, 04620, Republic of Korea
| | - Siyoung Kim
- Department of Biomedical Engineering, Dongguk University, Pildong-ro 1-gil 30, Jung-gu, Seoul, 04620, Republic of Korea
| | - Jongpil Kim
- Department of Biomedical Engineering, Dongguk University, Pildong-ro 1-gil 30, Jung-gu, Seoul, 04620, Republic of Korea. .,Department of Chemistry, Dongguk University, 30, Pildong-ro 1-gil 30, Jung-gu, Seoul, 04620, Republic of Korea.
| |
Collapse
|
34
|
Hu Y, Zhang F, Zhong W, Liu Y, He Q, Yang M, Chen H, Xu X, Bian K, Xu J, Li J, Shen Y, Zhang H. Transplantation of neural scaffolds consisting of dermal fibroblast-reprogrammed neurons and 3D silk fibrous materials promotes the repair of spinal cord injury. J Mater Chem B 2019; 7:7525-7539. [PMID: 31720683 DOI: 10.1039/c9tb01929d] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Neural scaffolds consisting of dermal fibroblast-reprogrammed neurons and 3D silk fibrous materials promote repair of spinal cord injury.
Collapse
|
35
|
Tsifaki M, Kelaini S, Caines R, Yang C, Margariti A. Regenerating the Cardiovascular System Through Cell Reprogramming; Current Approaches and a Look Into the Future. Front Cardiovasc Med 2018; 5:109. [PMID: 30177971 PMCID: PMC6109758 DOI: 10.3389/fcvm.2018.00109] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 07/24/2018] [Indexed: 12/19/2022] Open
Abstract
Cardiovascular disease (CVD), despite the advances of the medical field, remains one of the leading causes of mortality worldwide. Discovering novel treatments based on cell therapy or drugs is critical, and induced pluripotent stem cells (iPS Cells) technology has made it possible to design extensive disease-specific in vitro models. Elucidating the differentiation process challenged our previous knowledge of cell plasticity and capabilities and allows the concept of cell reprogramming technology to be established, which has inspired the creation of both in vitro and in vivo techniques. Patient-specific cell lines provide the opportunity of studying their pathophysiology in vitro, which can lead to novel drug development. At the same time, in vivo models have been designed where in situ transdifferentiation of cell populations into cardiomyocytes or endothelial cells (ECs) give hope toward effective cell therapies. Unfortunately, the efficiency as well as the concerns about the safety of all these methods make it exceedingly difficult to pass to the clinical trial phase. It is our opinion that creating an ex vivo model out of patient-specific cells will be one of the most important goals in the future to help surpass all these hindrances. Thus, in this review we aim to present the current state of research in reprogramming toward the cardiovascular system's regeneration, and showcase how the development and study of a multicellular 3D ex vivo model will improve our fighting chances.
Collapse
Affiliation(s)
- Marianna Tsifaki
- The Wellcome-Wolfson Building, Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Sophia Kelaini
- The Wellcome-Wolfson Building, Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Rachel Caines
- The Wellcome-Wolfson Building, Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Chunbo Yang
- The Wellcome-Wolfson Building, Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Andriana Margariti
- The Wellcome-Wolfson Building, Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| |
Collapse
|