1
|
Gong W, Dsouza N, Garry DJ. SeATAC: a tool for exploring the chromatin landscape and the role of pioneer factors. Genome Biol 2023; 24:125. [PMID: 37218013 DOI: 10.1186/s13059-023-02954-5] [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: 04/26/2022] [Accepted: 04/27/2023] [Indexed: 05/24/2023] Open
Abstract
Assay for Transposase-Accessible Chromatin with sequencing (ATAC-seq) reveals chromatin accessibility across the genome. Currently, no method specifically detects differential chromatin accessibility. Here, SeATAC uses a conditional variational autoencoder model to learn the latent representation of ATAC-seq V-plots and outperforms MACS2 and NucleoATAC on six separate tasks. Applying SeATAC to several pioneer factor-induced differentiation or reprogramming ATAC-seq datasets suggests that induction of these factors not only relaxes the closed chromatin but also decreases chromatin accessibility of 20% to 30% of their target sites. SeATAC is a novel tool to accurately reveal genomic regions with differential chromatin accessibility from ATAC-seq data.
Collapse
Affiliation(s)
- Wuming Gong
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN, 55455, USA.
- Lillehei Heart Institute, University of Minnesota, 2231 6Th St SE, Minneapolis, MN, 55455, USA.
| | - Nikita Dsouza
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Daniel J Garry
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN, 55455, USA.
- Lillehei Heart Institute, University of Minnesota, 2231 6Th St SE, Minneapolis, MN, 55455, USA.
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, 55455, USA.
- Paul and Sheila Wellstone Muscular Dystrophy Center, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
2
|
Herdy JR, Traxler L, Agarwal RK, Karbacher L, Schlachetzki JCM, Boehnke L, Zangwill D, Galasko D, Glass CK, Mertens J, Gage FH. Increased post-mitotic senescence in aged human neurons is a pathological feature of Alzheimer's disease. Cell Stem Cell 2022; 29:1637-1652.e6. [PMID: 36459967 PMCID: PMC10093780 DOI: 10.1016/j.stem.2022.11.010] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 10/03/2022] [Accepted: 11/09/2022] [Indexed: 12/02/2022]
Abstract
The concept of senescence as a phenomenon limited to proliferating cells has been challenged by growing evidence of senescence-like features in terminally differentiated cells, including neurons. The persistence of senescent cells late in life is associated with tissue dysfunction and increased risk of age-related disease. We found that Alzheimer's disease (AD) brains have significantly higher proportions of neurons that express senescence markers, and their distribution indicates bystander effects. AD patient-derived directly induced neurons (iNs) exhibit strong transcriptomic, epigenetic, and molecular biomarker signatures, indicating a specific human neuronal senescence-like state. AD iN single-cell transcriptomics revealed that senescent-like neurons face oncogenic challenges and metabolic dysfunction as well as display a pro-inflammatory signature. Integrative profiling of the inflammatory secretome of AD iNs and patient cerebral spinal fluid revealed a neuronal senescence-associated secretory phenotype that could trigger astrogliosis in human astrocytes. Finally, we show that targeting senescence-like neurons with senotherapeutics could be a strategy for preventing or treating AD.
Collapse
Affiliation(s)
- Joseph R Herdy
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA; Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA; Neural Aging Laboratory, Institute of Molecular Biology, CMBI, Leopold-Franzens-University Innsbruck, Tyrol, Austria.
| | - Larissa Traxler
- Neural Aging Laboratory, Institute of Molecular Biology, CMBI, Leopold-Franzens-University Innsbruck, Tyrol, Austria
| | - Ravi K Agarwal
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Lukas Karbacher
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Johannes C M Schlachetzki
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Lena Boehnke
- Neural Aging Laboratory, Institute of Molecular Biology, CMBI, Leopold-Franzens-University Innsbruck, Tyrol, Austria
| | - Dina Zangwill
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA; Division of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Doug Galasko
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Jerome Mertens
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA; Neural Aging Laboratory, Institute of Molecular Biology, CMBI, Leopold-Franzens-University Innsbruck, Tyrol, Austria.
| | - Fred H Gage
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA.
| |
Collapse
|
3
|
Guo Y, Wang YY, Sun TT, Xu JJ, Yang P, Ma CY, Guan WJ, Wang CJ, Liu GF, Liu CQ. Neural progenitor cells derived from fibroblasts induced by small molecule compounds under hypoxia for treatment of Parkinson's disease in rats. Neural Regen Res 2022; 18:1090-1098. [PMID: 36254998 PMCID: PMC9827776 DOI: 10.4103/1673-5374.355820] [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] [Indexed: 11/05/2022] Open
Abstract
Neural progenitor cells (NPCs) capable of self-renewal and differentiation into neural cell lineages offer broad prospects for cell therapy for neurodegenerative diseases. However, cell therapy based on NPC transplantation is limited by the inability to acquire sufficient quantities of NPCs. Previous studies have found that a chemical cocktail of valproic acid, CHIR99021, and Repsox (VCR) promotes mouse fibroblasts to differentiate into NPCs under hypoxic conditions. Therefore, we used VCR (0.5 mM valproic acid, 3 μM CHIR99021, and 1 μM Repsox) to induce the reprogramming of rat embryonic fibroblasts into NPCs under a hypoxic condition (5%). These NPCs exhibited typical neurosphere-like structures that can express NPC markers, such as Nestin, SRY-box transcription factor 2, and paired box 6 (Pax6), and could also differentiate into multiple types of functional neurons and astrocytes in vitro. They had similar gene expression profiles to those of rat brain-derived neural stem cells. Subsequently, the chemically-induced NPCs (ciNPCs) were stereotactically transplanted into the substantia nigra of 6-hydroxydopamine-lesioned parkinsonian rats. We found that the ciNPCs exhibited long-term survival, migrated long distances, and differentiated into multiple types of functional neurons and glial cells in vivo. Moreover, the parkinsonian behavioral defects of the parkinsonian model rats grafted with ciNPCs showed remarkable functional recovery. These findings suggest that rat fibroblasts can be directly transformed into NPCs using a chemical cocktail of VCR without introducing exogenous factors, which may be an attractive donor material for transplantation therapy for Parkinson's disease.
Collapse
Affiliation(s)
- Yu Guo
- School of Laboratory Medicine, School of Life Sciences, Bengbu Medical College, Bengbu, Anhui Province, China
| | - Yuan-Yuan Wang
- School of Laboratory Medicine, School of Life Sciences, Bengbu Medical College, Bengbu, Anhui Province, China
| | - Ting-Ting Sun
- School of Laboratory Medicine, School of Life Sciences, Bengbu Medical College, Bengbu, Anhui Province, China
| | - Jia-Jia Xu
- School of Laboratory Medicine, School of Life Sciences, Bengbu Medical College, Bengbu, Anhui Province, China
| | - Pan Yang
- School of Laboratory Medicine, School of Life Sciences, Bengbu Medical College, Bengbu, Anhui Province, China
| | - Cai-Yun Ma
- School of Laboratory Medicine, School of Life Sciences, Bengbu Medical College, Bengbu, Anhui Province, China,National Germplasm Resource Center for Domestic Animals, Institute of Animal Science, Chinese Academy of Agricultural Science, Beijing, China
| | - Wei-Jun Guan
- National Germplasm Resource Center for Domestic Animals, Institute of Animal Science, Chinese Academy of Agricultural Science, Beijing, China
| | - Chun-Jing Wang
- School of Laboratory Medicine, School of Life Sciences, Bengbu Medical College, Bengbu, Anhui Province, China
| | - Gao-Feng Liu
- School of Laboratory Medicine, School of Life Sciences, Bengbu Medical College, Bengbu, Anhui Province, China,Correspondence to: Chang-Qing Liu, ; Gao-Feng Liu, .
| | - Chang-Qing Liu
- School of Laboratory Medicine, School of Life Sciences, Bengbu Medical College, Bengbu, Anhui Province, China,Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, USA,Correspondence to: Chang-Qing Liu, ; Gao-Feng Liu, .
| |
Collapse
|
4
|
Pan Y, Gu Z, Lyu Y, Yang Y, Chung M, Pan X, Cai S. Link between senescence and cell fate: Senescence-associated secretory phenotype (SASP) and its effects on stem cell fate transition. Rejuvenation Res 2022; 25:160-172. [PMID: 35658548 DOI: 10.1089/rej.2022.0021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Senescence is a form of durable cell cycle arrest elicited in response to a wide range of stimuli. Senescent cells remain metabolically active and secrete a variety of factors collectively termed senescence-associated secretory phenotype (SASP). SASP is highly pleiotropic and can impact numerous biological processes in which it has both beneficial and deleterious roles. The underlying mechanisms by which SASP exerts its pleiotropic influence remain largely unknown. SASP serves as an environmental factor, which regulates stem cell differentiation and alters its routine. The latter can potentially be accomplished through dedifferentiation, transdifferentiation, or reprogramming. Behavioral changes that cells undergo when exposed to SASP are involved in several senescence-associated physiological and pathological phenomena. These findings provide clues for identifying possible interventions to reduce the deleterious effects without interfering in the beneficial outcomes. Here, we discuss the multifaced effects of SASP and the changes occurring in cellular states upon exposure to SASP factors.
Collapse
Affiliation(s)
- Yu Pan
- Shenzhen University, 47890, Shenzhen, Guangdong, China;
| | - Zhenzhen Gu
- Shenzhen University, 47890, Shenzhen, Guangdong, China;
| | - Yansi Lyu
- Shenzhen University, 47890, Shenzhen, Guangdong, China;
| | - Yi Yang
- Shenzhen University, 47890, Shenzhen, Guangdong, China;
| | - Manhon Chung
- Shanghai Jiao Tong University School of Medicine, 56694, Shanghai, China;
| | - Xiaohua Pan
- Shenzhen University, 47890, Shenzhen, Guangdong, China;
| | - Sa Cai
- Shenzhen University, 47890, 3688 Nanhai Avenue, Nanshan District, Shenzhen, Shenzhen, China, 518060;
| |
Collapse
|
5
|
Bindra D, Mishra RK. In Pursuit of Distinctiveness: Transmembrane Nucleoporins and Their Disease Associations. Front Oncol 2022; 11:784319. [PMID: 34970494 PMCID: PMC8712647 DOI: 10.3389/fonc.2021.784319] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 11/25/2021] [Indexed: 11/13/2022] Open
Abstract
The bi-directional nucleocytoplasmic shuttling of macromolecules like molecular signals, transcription factors, regulatory proteins, and RNAs occurs exclusively through Nuclear Pore Complex (NPC) residing in the nuclear membrane. This magnanimous complex is essentially a congregation of ~32 conserved proteins termed Nucleoporins (Nups) present in multiple copies and mostly arranged as subcomplexes to constitute a functional NPC. Nups participate in ancillary functions such as chromatin organization, transcription regulation, DNA damage repair, genome stabilization, and cell cycle control, apart from their central role as nucleocytoplasmic conduits. Thus, Nups exert a role in the maintenance of cellular homeostasis. In mammals, precisely three nucleoporins traverse the nuclear membrane, are called transmembrane Nups (TM-Nups), and are involved in multiple cellular functions. Owing to their vital roles in cellular processes and homeostasis, dysregulation of nucleoporin function is implicated in various diseases. The deregulated functioning of TM-Nups can thus act as an opportune window for the development of diseases. Indeed, mounting evidence exhibits a strong association of TM-Nups in cancer and numerous other physiological disorders. These findings have provided much-needed insights into the novel mechanisms of disease progression. While nucleoporin’s functions have often been summarized in the disease context, a focus on TM-Nups has always lacked. This review emphasizes the elucidation of distinct canonical and non-canonical functions of mammalian TM-Nups and the underlying mechanisms of their disease association.
Collapse
Affiliation(s)
- Divya Bindra
- Nups and SUMO Biology Group, Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Bhopal, Bhopal, India
| | - Ram Kumar Mishra
- Nups and SUMO Biology Group, Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Bhopal, Bhopal, India
| |
Collapse
|
6
|
Fernandez-Muñoz B, Garcia-Delgado AB, Arribas-Arribas B, Sanchez-Pernaute R. Human Neural Stem Cells for Cell-Based Medicinal Products. Cells 2021; 10:2377. [PMID: 34572024 PMCID: PMC8469920 DOI: 10.3390/cells10092377] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 12/15/2022] Open
Abstract
Neural stem cells represent an attractive tool for the development of regenerative therapies and are being tested in clinical trials for several neurological disorders. Human neural stem cells can be isolated from the central nervous system or can be derived in vitro from pluripotent stem cells. Embryonic sources are ethically controversial and other sources are less well characterized and/or inefficient. Recently, isolation of NSC from the cerebrospinal fluid of patients with spina bifida and with intracerebroventricular hemorrhage has been reported. Direct reprogramming may become another alternative if genetic and phenotypic stability of the reprogrammed cells is ensured. Here, we discuss the advantages and disadvantages of available sources of neural stem cells for the production of cell-based therapies for clinical applications. We review available safety and efficacy clinical data and discuss scalability and quality control considerations for manufacturing clinical grade cell products for successful clinical application.
Collapse
Affiliation(s)
- Beatriz Fernandez-Muñoz
- Cellular Reprogramming and Production Unit, Andalusian Network for the Design and Translation of Advanced Therapies, 41092 Sevilla, Spain; (A.B.G.-D.); (B.A.-A.)
| | - Ana Belen Garcia-Delgado
- Cellular Reprogramming and Production Unit, Andalusian Network for the Design and Translation of Advanced Therapies, 41092 Sevilla, Spain; (A.B.G.-D.); (B.A.-A.)
| | - Blanca Arribas-Arribas
- Cellular Reprogramming and Production Unit, Andalusian Network for the Design and Translation of Advanced Therapies, 41092 Sevilla, Spain; (A.B.G.-D.); (B.A.-A.)
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Sevilla, 41012 Sevilla, Spain
| | - Rosario Sanchez-Pernaute
- Cellular Reprogramming and Production Unit, Andalusian Network for the Design and Translation of Advanced Therapies, 41092 Sevilla, Spain; (A.B.G.-D.); (B.A.-A.)
| |
Collapse
|
7
|
Abstract
Nuclear pore complexes are multiprotein channels that span the nuclear envelope, which connects the nucleus to the cytoplasm. In addition to their main role in the regulation of nucleocytoplasmic molecule exchange, it has become evident that nuclear pore complexes and their components also have multiple transport-independent functions. In recent years, an increasing number of studies have reported the involvement of nuclear pore complex components in embryogenesis, cell differentiation and tissue-specific processes. Here, we review the findings that highlight the dynamic nature of nuclear pore complexes and their roles in many cell type-specific functions during development and tissue homeostasis.
Collapse
Affiliation(s)
- Valeria Guglielmi
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
- NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | | | - Maximiliano A D'Angelo
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
- NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| |
Collapse
|
8
|
Wei C, Yu P, Cheng L. Hematopoietic Reprogramming Entangles with Hematopoiesis. Trends Cell Biol 2020; 30:752-763. [PMID: 32861580 DOI: 10.1016/j.tcb.2020.07.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/24/2020] [Accepted: 07/28/2020] [Indexed: 02/06/2023]
Abstract
Hematopoiesis generally refers to hematopoietic development in fetuses and adults, as well as to hematopoietic stem cell differentiation into progeny lineages. The multiple processes that generate diverse hematopoietic cells have been considered to be unidirectional. However, many reports have recently demonstrated that these processes are not only reversible but also interconvertible via cell reprogramming. The cell reprogramming that occurs in hematopoietic cells is termed hematopoietic reprogramming. We focus on both autogenous and artificial hematopoietic reprogramming under physiological and pathological conditions that is mainly directed by the actions of transcription factors (TFs), chemical compounds, or extracellular cytokines. A comprehensive understanding of hematopoietic reprogramming will help us not only to generate desirable cells for cell therapy but also to further analyze normal and malignant hematopoiesis.
Collapse
Affiliation(s)
- Chuijin Wei
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Pei Yu
- Department of Orthopaedics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Lin Cheng
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
9
|
Wang J, Jiang X, Zhao L, Zuo S, Chen X, Zhang L, Lin Z, Zhao X, Qin Y, Zhou X, Yu XY. Lineage reprogramming of fibroblasts into induced cardiac progenitor cells by CRISPR/Cas9-based transcriptional activators. Acta Pharm Sin B 2020; 10:313-326. [PMID: 32082976 PMCID: PMC7016296 DOI: 10.1016/j.apsb.2019.09.003] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 08/26/2019] [Accepted: 08/29/2019] [Indexed: 12/16/2022] Open
Abstract
Overexpression of exogenous lineage-determining factors succeeds in directly reprogramming fibroblasts to various cell types. Several studies have reported reprogramming of fibroblasts into induced cardiac progenitor cells (iCPCs). CRISPR/Cas9-mediated gene activation is a potential approach for cellular reprogramming due to its high precision and multiplexing capacity. Here we show lineage reprogramming to iCPCs through a dead Cas9 (dCas9)-based transcription activation system. Targeted and robust activation of endogenous cardiac factors, including GATA4, HAND2, MEF2C and TBX5 (G, H, M and T; GHMT), can reprogram human fibroblasts toward iCPCs. The iCPCs show potentials to differentiate into cardiomyocytes, smooth muscle cells and endothelial cells in vitro. Addition of MEIS1 to GHMT induces cell cycle arrest in G2/M and facilitates cardiac reprogramming. Lineage reprogramming of human fibroblasts into iCPCs provides a promising cellular resource for disease modeling, drug discovery and individualized cardiac cell therapy.
Collapse
|
10
|
Reprogramming of Fibroblasts to Neural Stem Cells by a Chemical Cocktail. Methods Mol Biol 2020. [PMID: 31960385 DOI: 10.1007/978-1-0716-0301-7_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Chemically induced cell fate conversion, including reprogramming to pluripotent stem cells and direct reprogramming to somatic cells, has been proved to be an alternative strategy with many advantages, in comparison with conventional transcription factors- or microRNAs-enabled cell reprogramming. Many functional and desirable cells have been generated via the chemically induced reprogramming. Neural stem cells (NSCs) hold great potential in basic research and clinical application. Here, we describe a detailed protocol for converting mouse fibroblasts into NSCs by a cocktail of chemical compounds.
Collapse
|
11
|
Zhou Y, Zhu X, Dai Y, Xiong S, Wei C, Yu P, Tang Y, Wu L, Li J, Liu D, Wang Y, Chen Z, Chen S, Huang J, Cheng L. Chemical Cocktail Induces Hematopoietic Reprogramming and Expands Hematopoietic Stem/Progenitor Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1901785. [PMID: 31921559 PMCID: PMC6947705 DOI: 10.1002/advs.201901785] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 10/20/2019] [Indexed: 05/03/2023]
Abstract
Generation of hematopoietic stem/progenitor cells (HSPCs) via cell expansion or cell reprogramming has been widely achieved by overexpression of transcription factors. Herein, it is reported that without introducing exogenous genes, mouse fibroblasts can be reprogrammed into hemogenic cells based on lineage tracing analysis, which further develop into hematopoietic cells, by treatment of cocktails of chemical compounds. The chemical cocktails also reprogram differentiated hematopoietic cells back into HSPC-like cells. Most importantly, the chemical cocktails enabling hematopoietic reprogramming robustly promote HSPC proliferation ex vivo. The expanded HSPCs acquire enhanced capacity of hematopoietic reconstruction in vivo. Single-cell sequencing analysis verifies the expansion of HSPCs and the cell reprogramming toward potential generation of HSPCs at the same time by the chemical cocktail treatment. Thus, the proof-of-concept findings not only demonstrate that hematopoietic reprogramming can be achieved by chemical compounds but also provide a promising strategy for acquisition of HSPCs by chemical cocktail-enabled double effects.
Collapse
Affiliation(s)
- Yi Zhou
- State Key Laboratory of Medical GenomicsShanghai Institute of HematologyRui Jin Hospital affiliated to Shanghai Jiao Tong UniversitySchool of MedicineKey Laboratory of Systems Biomedicine (Ministry of Education) and Collaborative Innovation Center of Systems BiomedicineShanghai Center for Systems BiomedicineShanghai Jiao Tong UniversityShanghai200025China
| | - Xingli Zhu
- State Key Laboratory of Medical GenomicsShanghai Institute of HematologyRui Jin Hospital affiliated to Shanghai Jiao Tong UniversitySchool of MedicineKey Laboratory of Systems Biomedicine (Ministry of Education) and Collaborative Innovation Center of Systems BiomedicineShanghai Center for Systems BiomedicineShanghai Jiao Tong UniversityShanghai200025China
| | - Yuting Dai
- School of Life Sciences and BiotechnologyShanghai Jiao Tong University200025ShanghaiChina
| | - Shumin Xiong
- State Key Laboratory of Medical GenomicsShanghai Institute of HematologyRui Jin Hospital affiliated to Shanghai Jiao Tong UniversitySchool of MedicineKey Laboratory of Systems Biomedicine (Ministry of Education) and Collaborative Innovation Center of Systems BiomedicineShanghai Center for Systems BiomedicineShanghai Jiao Tong UniversityShanghai200025China
| | - Chuijin Wei
- State Key Laboratory of Medical GenomicsShanghai Institute of HematologyRui Jin Hospital affiliated to Shanghai Jiao Tong UniversitySchool of MedicineKey Laboratory of Systems Biomedicine (Ministry of Education) and Collaborative Innovation Center of Systems BiomedicineShanghai Center for Systems BiomedicineShanghai Jiao Tong UniversityShanghai200025China
| | - Pei Yu
- Department of OrthopaedicsRui Jin Hospital affiliated to Shanghai Jiao Tong University School of MedicineShanghai200025China
| | - Yuewen Tang
- State Key Laboratory of Medical GenomicsShanghai Institute of HematologyRui Jin Hospital affiliated to Shanghai Jiao Tong UniversitySchool of MedicineKey Laboratory of Systems Biomedicine (Ministry of Education) and Collaborative Innovation Center of Systems BiomedicineShanghai Center for Systems BiomedicineShanghai Jiao Tong UniversityShanghai200025China
| | - Liang Wu
- State Key Laboratory of Medical GenomicsShanghai Institute of HematologyRui Jin Hospital affiliated to Shanghai Jiao Tong UniversitySchool of MedicineKey Laboratory of Systems Biomedicine (Ministry of Education) and Collaborative Innovation Center of Systems BiomedicineShanghai Center for Systems BiomedicineShanghai Jiao Tong UniversityShanghai200025China
| | - Jianfeng Li
- State Key Laboratory of Medical GenomicsShanghai Institute of HematologyRui Jin Hospital affiliated to Shanghai Jiao Tong UniversitySchool of MedicineKey Laboratory of Systems Biomedicine (Ministry of Education) and Collaborative Innovation Center of Systems BiomedicineShanghai Center for Systems BiomedicineShanghai Jiao Tong UniversityShanghai200025China
| | - Dan Liu
- State Key Laboratory of Medical GenomicsShanghai Institute of HematologyRui Jin Hospital affiliated to Shanghai Jiao Tong UniversitySchool of MedicineKey Laboratory of Systems Biomedicine (Ministry of Education) and Collaborative Innovation Center of Systems BiomedicineShanghai Center for Systems BiomedicineShanghai Jiao Tong UniversityShanghai200025China
| | - Yanlin Wang
- Prenatal Diagnosis CenterInternational Peace Maternity and Child Health Hospital affiliated to Shanghai Jiao Tong University School of MedicineShanghai200030China
| | - Zhu Chen
- State Key Laboratory of Medical GenomicsShanghai Institute of HematologyRui Jin Hospital affiliated to Shanghai Jiao Tong UniversitySchool of MedicineKey Laboratory of Systems Biomedicine (Ministry of Education) and Collaborative Innovation Center of Systems BiomedicineShanghai Center for Systems BiomedicineShanghai Jiao Tong UniversityShanghai200025China
| | - Sai‐Juan Chen
- State Key Laboratory of Medical GenomicsShanghai Institute of HematologyRui Jin Hospital affiliated to Shanghai Jiao Tong UniversitySchool of MedicineKey Laboratory of Systems Biomedicine (Ministry of Education) and Collaborative Innovation Center of Systems BiomedicineShanghai Center for Systems BiomedicineShanghai Jiao Tong UniversityShanghai200025China
| | - Jinyan Huang
- State Key Laboratory of Medical GenomicsShanghai Institute of HematologyRui Jin Hospital affiliated to Shanghai Jiao Tong UniversitySchool of MedicineKey Laboratory of Systems Biomedicine (Ministry of Education) and Collaborative Innovation Center of Systems BiomedicineShanghai Center for Systems BiomedicineShanghai Jiao Tong UniversityShanghai200025China
- Pôle de Recherches Sino‐Français en Science du Vivant et GénomiqueLaboratory of Molecular PathologyRui‐Jin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Lin Cheng
- State Key Laboratory of Medical GenomicsShanghai Institute of HematologyRui Jin Hospital affiliated to Shanghai Jiao Tong UniversitySchool of MedicineKey Laboratory of Systems Biomedicine (Ministry of Education) and Collaborative Innovation Center of Systems BiomedicineShanghai Center for Systems BiomedicineShanghai Jiao Tong UniversityShanghai200025China
| |
Collapse
|
12
|
Disease-specific alteration of karyopherin-α subtype establishes feed-forward oncogenic signaling in head and neck squamous cell carcinoma. Oncogene 2019; 39:2212-2223. [PMID: 31822798 PMCID: PMC7056645 DOI: 10.1038/s41388-019-1137-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 11/25/2019] [Accepted: 11/27/2019] [Indexed: 12/11/2022]
Abstract
Nuclear import, mediated in part by karyopherin-α (KPNA)/importin-α subtypes, regulates transcription factor access to the genome and determines cell fate. However, the cancer-specific changes of KPNA subtypes and the relevancy in cancer biology remain largely unknown. Here, we report that KPNA4, encoding karyopherin-α4 (KPNA4), is exclusively amplified and overexpressed in head and neck of squamous cell carcinoma (HNSCC). Depletion of KPNA4 attenuated nuclear localization signal-dependent transport activity and suppressed malignant phenotypes and induced epidermal differentiation. Mechanistically, KPNA4-mediated nuclear transport of Ras-responsive element-binding protein (RREB1), which sustains Ras/ERK pathway signaling through repressing miR-143/145 expression. Notably, MAPK signaling enhanced trafficking activity of KPNA4 via phosphorylation of KPNA4 at Ser60. These data reveal that KPNA4 establishes a feed-forward cascade that potentiates Ras/ERK signaling in HNSCC.
Collapse
|
13
|
Erharter A, Rizzi S, Mertens J, Edenhofer F. Take the shortcut - direct conversion of somatic cells into induced neural stem cells and their biomedical applications. FEBS Lett 2019; 593:3353-3369. [PMID: 31663609 PMCID: PMC6916337 DOI: 10.1002/1873-3468.13656] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 10/12/2019] [Accepted: 10/14/2019] [Indexed: 12/20/2022]
Abstract
Second-generation reprogramming of somatic cells directly into the cell type of interest avoids induction of pluripotency and subsequent cumbersome differentiation procedures. Several recent studies have reported direct conversion of human somatic cells into stably proliferating induced neural stem cells (iNSCs). Importantly, iNSCs are easier, faster, and more cost-efficient to generate than induced pluripotent stem cells (iPSCs), and also have a higher level of clinical safety. Stably, self-renewing iNSCs can be derived from different cellular sources, such as skin fibroblasts and peripheral blood mononuclear cells, and readily differentiate into neuronal and glial lineages that are indistinguishable from their iPSC-derived counterparts or from NSCs isolated from primary tissues. This review focuses on the derivation and characterization of iNSCs and their biomedical applications. We first outline different approaches to generate iNSCs and then discuss the underlying molecular mechanisms. Finally, we summarize the preclinical validation of iNSCs to highlight that these cells are promising targets for disease modeling, autologous cell therapy, and precision medicine.
Collapse
Affiliation(s)
- Anita Erharter
- Department of Molecular Biology & CMBIGenomics, Stem Cell Biology & Regenerative MedicineLeopold‐Franzens‐University InnsbruckAustria
| | - Sandra Rizzi
- Department of Molecular Biology & CMBIGenomics, Stem Cell Biology & Regenerative MedicineLeopold‐Franzens‐University InnsbruckAustria
- Institute of PharmacologyMedical University InnsbruckAustria
| | - Jerome Mertens
- Department of Molecular Biology & CMBIGenomics, Stem Cell Biology & Regenerative MedicineLeopold‐Franzens‐University InnsbruckAustria
| | - Frank Edenhofer
- Department of Molecular Biology & CMBIGenomics, Stem Cell Biology & Regenerative MedicineLeopold‐Franzens‐University InnsbruckAustria
| |
Collapse
|
14
|
Cellular Reprogramming as a Therapeutic Target in Cancer. Trends Cell Biol 2019; 29:623-634. [PMID: 31153655 DOI: 10.1016/j.tcb.2019.05.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 04/28/2019] [Accepted: 05/01/2019] [Indexed: 12/30/2022]
Abstract
Cancer heterogeneity has long been recognized as an important clinical determinant of patient outcomes and, thus, many new cancer treatments have been designed to target these different cells. Despite the short-term achievements of current therapies, including chemotherapy, antiangiogenesis therapy, radiotherapy, and immunotherapy, the long-term success of cancer regression remains poor. Therefore, researchers have investigated a new property, cellular reprogramming, in cancer that not only contributes to the classic hallmarks of cancer, but also suggests that cancer is a dynamic event rather than a static cellular entity. Here, we discuss the mechanisms by which the cellular reprogramming of cancer cells can explain some of the phenotypic and functional heterogeneity observed among cancer cells.
Collapse
|