1
|
Nguyen LD, Sengupta S, Cho KI, Floru A, George RE, Krichevsky AM. A drug that induces the microRNA miR-124 enables differentiation of retinoic acid-resistant neuroblastoma cells. Sci Signal 2025; 18:eads2641. [PMID: 40233178 DOI: 10.1126/scisignal.ads2641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 03/27/2025] [Indexed: 04/17/2025]
Abstract
Tumor cell heterogeneity in neuroblastoma, a pediatric cancer arising from neural crest-derived progenitor cells, presents clinical challenges. Unlike adrenergic (ADRN) neuroblastoma cells, neuroblastoma cells with a mesenchymal (MES) identity are resistant to chemotherapy and retinoid therapy, which contributes to relapses and treatment failures. We explored whether up-regulation of the neurogenic, tumor suppressor microRNA miR-124 could promote the differentiation of retinoic acid-resistant MES neuroblastoma cells. Leveraging our screen for miRNA-modulatory small molecules, we identified and validated the tyrosine and phosphoinositide kinase inhibitor PP121 as a robust inducer of miR-124. Combining PP121 and BDNF-activating bufalin synergistically arrested proliferation and promoted the sustained differentiation of MES/heterogeneous SK-N-AS cells over several weeks. This protocol also resulted in the differentiation of multiple MES neuroblastoma and glioblastoma cell lines. RNA-seq analysis of differentiated MES/heterogeneous SK-N-AS cells revealed the replacement of the ADRN core regulatory circuitry with circuitries associated with chromaffin cells and Schwann cell precursors. Furthermore, differentiation was associated with inhibition of the CDK4/CDK6 pathway and activation of a transcriptional program that correlated with improved outcomes for patients with neuroblastoma. Our findings suggest an approach with translational potential to induce the differentiation of therapy-resistant cancers of the nervous system. Moreover, these long-lived, differentiated cells could be used to study mechanisms underlying cancer biology and therapies.
Collapse
Affiliation(s)
- Lien D Nguyen
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Satyaki Sengupta
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, 02115, USA
| | - Kevin I Cho
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Alexander Floru
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, 02115, USA
| | - Rani E George
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, 02115, USA
| | - Anna M Krichevsky
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
2
|
Vrancx C, Annaert W. Lysosome repair fails in ageing and Alzheimer's disease. Nat Cell Biol 2025; 27:553-555. [PMID: 40140604 DOI: 10.1038/s41556-024-01608-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Affiliation(s)
- Céline Vrancx
- Laboratory for Membrane Trafficking, VIB Center for Brain and Disease Research, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Wim Annaert
- Laboratory for Membrane Trafficking, VIB Center for Brain and Disease Research, Leuven, Belgium.
- Department of Neurosciences, KU Leuven, Leuven, Belgium.
| |
Collapse
|
3
|
Li Y, Sun J, Xu T, Dai B, Wang Y. Efficient and rapid generation of neural stem cells by direct conversion of fibroblasts with single microRNAs. Stem Cells 2025; 43:sxaf003. [PMID: 39862169 DOI: 10.1093/stmcls/sxaf003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/06/2025] [Indexed: 01/27/2025]
Abstract
Neural stem cells (NSCs) hold great potential in neurodegenerative disease therapy, drug screening, and disease modeling. However, current approaches for induced NSCs (iNSCs) generation from somatic cells are still slow and inefficient. Here we report the establishment of a rapid and efficient method of iNSCs generation from human and mouse fibroblasts by using single microRNAs (miR-302a). These iNSCs exhibited morphological, molecular and functional properties resembling those of adult human and mouse NSCs, respectively. Additionally, human iNSCs can be expanded for more than 20 passages in vitro. Furthermore, miR-302a alone was demonstrated to be sufficient to reprogram both human and mouse fibroblasts into iNSCs. Our results showed a method of direct conversion of autologous fibroblasts with miR-302a into iNSCs, providing a rapid and efficient strategy to generate iNSCs for both basic research and clinical applications.
Collapse
Affiliation(s)
- Yuanyuan Li
- Medicine and Pharmacy Research Center, and Yantai Key Laboratory for Stem Cell Biology and Regenerative Medicine, Binzhou Medical University, Yantai, Shandong 264003, China
| | - Jing Sun
- Medicine and Pharmacy Research Center, and Yantai Key Laboratory for Stem Cell Biology and Regenerative Medicine, Binzhou Medical University, Yantai, Shandong 264003, China
| | - Tingting Xu
- Medicine and Pharmacy Research Center, and Yantai Key Laboratory for Stem Cell Biology and Regenerative Medicine, Binzhou Medical University, Yantai, Shandong 264003, China
| | - Bo Dai
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Yuesi Wang
- Medicine and Pharmacy Research Center, and Yantai Key Laboratory for Stem Cell Biology and Regenerative Medicine, Binzhou Medical University, Yantai, Shandong 264003, China
| |
Collapse
|
4
|
Woo E, Tasnim F, Kawamata H, Manfredi G, Konrad C. Investigation of mitochondrial phenotypes in motor neurons derived by direct conversion of fibroblasts from familial ALS subjects. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.13.637962. [PMID: 40027671 PMCID: PMC11870414 DOI: 10.1101/2025.02.13.637962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease of motor neurons, leading to fatal muscle paralysis. Familial forms of ALS (fALS) account for approximately 10% of cases and are associated with mutations in numerous genes. Alterations of mitochondrial functions have been proposed to contribute to disease pathogenesis. Here, we employed a direct conversion (DC) technique to generate induced motor neurons (iMN) from skin fibroblasts to investigate mitochondrial phenotypes in a patient-derived disease relevant cell culture system. We converted 7 control fibroblast lines and 17 lines harboring the following fALS mutations, SOD1 A4V , TDP-43 N352S , FUS R521G , CHCHD10 R15L , and C9orf72 repeat expansion. We developed new machine learning approaches to identify iMN, analyze their mitochondrial function, and follow their fate longitudinally. Mitochondrial and energetic abnormalities were observed, but not all fALS iMN lines exhibited the same alterations. SOD1 A4V , C9orf72, and TDP-43 N352S iMN had increased mitochondrial membrane potential, while in CHCHD10 R15L cells membrane potential was decreased. TDP-43 N352S iMN displayed changes in mitochondrial morphology and increased motility. SOD1 A4V , TDP-43 N352S , and CHCHD10 R15L iMN had increased oxygen consumption rates and altered extracellular acidification rates, reflecting a hypermetabolic state similar to the one described in sporadic ALS fibroblasts. FUS R521G mutants had decreased ATP/ADP ratio, suggesting impaired energy metabolism. We then tested the viability of iMN and found decreases in survival in SOD1 A4V , C9orf72, and FUS R521G , which were corrected by small molecules that target mitochondrial stress. Together, our findings reinforce the role of mitochondrial dysfunction in ALS and indicate that fibroblast-derived iMN may be useful to study fALS metabolic alterations. Strengths of the DC iMN approach include low cost, speed of transformation, and the preservation of epigenetic modifications. However, further refinement of the fibroblasts DC iMN technique is still needed to improve transformation efficiency, reproducibility, the relatively short lifespan of iMN, and the senescence of the parental fibroblasts.
Collapse
Affiliation(s)
- Evan Woo
- Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Faiza Tasnim
- Sophie Davis School of Biomedical Education, CUNY School of Medicine, New York, NY, USA
| | - Hibiki Kawamata
- Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Giovanni Manfredi
- Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Csaba Konrad
- Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
5
|
Burbach KF, Wu S, Yoo AS. Notch inhibition enhances morphological reprogramming of microRNA-induced human neurons. Stem Cells 2025; 43:sxae079. [PMID: 39573925 DOI: 10.1093/stmcls/sxae079] [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: 02/07/2024] [Accepted: 11/04/2024] [Indexed: 11/27/2024]
Abstract
The role of Notch signaling in direct neuronal reprogramming remains unknown despite its importance to brain development in vivo. Here, we use microRNA-induced neurons that are directly reprogrammed from human fibroblasts to determine how Notch signaling contributes to neuronal identity. We found that Notch inhibition during the first week of reprogramming was both necessary and sufficient to enhance neurite outgrowth at a later timepoint, indicating an important role in the erasure of the original cell identity. Accordingly, transcriptomic analysis showed that the effect of Notch inhibition was likely due to improvements in fibroblast fate erasure and silencing of non-neuronal genes. To this effect, we identify MYLIP, whose downregulation in response to Notch inhibition significantly promoted neurite outgrowth. Moreover, Notch inhibition resulted in cells with neuronal transcriptome signatures defined by expressing long genes at a faster rate than the control, demonstrating the effect of accelerated fate erasure on neuronal fate acquisition. Our results demonstrate the antagonistic role of Notch signaling to the pro-neuronal microRNAs 9 and 124 and the benefits of its inhibition to the acquisition of neuronal morphology.
Collapse
Affiliation(s)
- Kyle F Burbach
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, United States
- Program in Molecular Genetics and Genomics, Division of Biology & Biomedical Sciences, Washington University in St. Louis, St. Louis, MO 63110, United States
| | - Shanyun Wu
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, United States
- Program in Molecular Genetics and Genomics, Division of Biology & Biomedical Sciences, Washington University in St. Louis, St. Louis, MO 63110, United States
| | - Andrew S Yoo
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, United States
- Center for Regenerative Medicine, Washington University in St. Louis, St. Louis, MO 63110, United States
| |
Collapse
|
6
|
Cates K, Yuan L, Yang Y, Yoo AS. Fate erasure logic of gene networks underlying direct neuronal conversion of somatic cells by microRNAs. Cell Rep 2025; 44:115153. [PMID: 39756035 PMCID: PMC11834941 DOI: 10.1016/j.celrep.2024.115153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 09/10/2024] [Accepted: 12/13/2024] [Indexed: 01/07/2025] Open
Abstract
Neurogenic microRNAs 9/9∗ and 124 (miR-9/9∗-124) drive the direct reprogramming of human fibroblasts into neurons with the initiation of the fate erasure of fibroblasts. However, whether the miR-9/9∗-124 fate erasure logic extends to the neuronal conversion of other somatic cell types remains unknown. Here, we uncover that miR-9/9∗-124 induces neuronal conversion of multiple cell types: dura fibroblasts, astrocytes, smooth muscle cells, and pericytes. We reveal the cell-type-specific and pan-somatic gene network erasure induced by miR-9/9∗-124, including cell cycle, morphology, and proteostasis gene networks. Leveraging these pan-somatic gene networks, we predict upstream regulators that may antagonize somatic fate erasure. Among the predicted regulators, we identify TP53 (p53), whose inhibition is sufficient to enhance neuronal conversion even in post-mitotic cells. This study extends miR-9/9∗-124 reprogramming to alternate somatic cells, reveals the pan-somatic gene network fate erasure logic of miR-9/9∗-124, and shows a neurogenic role for p53 inhibition in the miR-9/9∗-124 signaling cascade.
Collapse
Affiliation(s)
- Kitra Cates
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Program in Molecular Genetics and Genomics, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Luorongxin Yuan
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Program in Molecular Genetics and Genomics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yan Yang
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Andrew S Yoo
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
7
|
Papadimitriou E, Koutsoudaki PN, Thanou I, Thomaidou D. In Vitro and In Vivo Direct Reprogramming of Astrocytes to Induced-Neurons. Methods Mol Biol 2025; 2899:183-198. [PMID: 40067625 DOI: 10.1007/978-1-0716-4386-0_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2025]
Abstract
Recent studies from a number of groups, including ours, have shown that astrocytes can be directly reprogrammed into induced neurons (iNs) in vitro and in vivo following ectopic overexpression of combinations of transcription factors (TFs), miRNAs, and small chemical molecules. Brain-enriched miRNAs in particular have been recently considered potent reprogramming factors due to their ability to post-transcriptionally modulate high numbers of neurogenic factors and have, thus, been introduced, supplementary or alternatively to TFs, to instruct direct neuronal reprogramming.In this chapter, we describe the methodology for in vitro direct reprogramming of primary mouse astrocytes to differentiated, functional iNs following forced expression of the neurogenic miRNA miR-124 along with the neuronal differentiation-promoting chemical compound Isoexasole-9 (ISX9). Further, we present the methodological procedure for in vivo converting resident activated cortical astrocytes to iNs, following stab wound injury of the mouse cortex and subsequent viral-mediated overexpression of miR-124 in the peritraumatic area along with ISX9 systemic administration.
Collapse
Affiliation(s)
- Elsa Papadimitriou
- Neural Stem Cells and Neuroimaging Group, Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| | - Paraskevi N Koutsoudaki
- Neural Stem Cells and Neuroimaging Group, Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Irini Thanou
- Neural Stem Cells and Neuroimaging Group, Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| | - Dimitra Thomaidou
- Neural Stem Cells and Neuroimaging Group, Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece.
| |
Collapse
|
8
|
Weigele J, Zhang L, Franco A, Cartier E, Dorn GW. Sensory-Motor Neuropathy in Mfn2 T105M Knock-in Mice and Its Reversal by a Novel Piperine-Derived Mitofusin Activator. J Pharmacol Exp Ther 2024; 391:361-374. [PMID: 39284622 PMCID: PMC11493442 DOI: 10.1124/jpet.124.002258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 08/09/2024] [Indexed: 10/20/2024] Open
Abstract
Mitochondrial dysfunction is a hallmark of many genetic neurodegenerative diseases, but therapeutic options to reverse mitochondrial dysfunction are limited. While recent studies support the possibility of improving mitochondrial fusion/fission dynamics and motility to correct mitochondrial dysfunction and resulting neurodegeneration in Charcot-Marie-Tooth disease (CMT) and other neuropathies, the clinical utility of reported compounds and relevance of preclinical models are uncertain. Here, we describe motor and sensory neuron dysfunction characteristic of clinical CMT type 2 A in a CRISPR/Casp-engineered Mfn2 Thr105Met (T105M) mutant knock-in mouse. We further demonstrate that daily oral treatment with a novel mitofusin activator derived from the natural product piperine can reverse these neurologic phenotypes. Piperine derivative 8015 promoted mitochondrial fusion and motility in Mfn2-deficient cells in a mitofusin-dependent manner and reversed mitochondrial dysfunction in cultured fibroblasts and reprogrammed motor neurons from a human CMT2A patient carrying the MFN2 T105M mutation. Like previous mitofusin activators, 8015 exhibited stereospecific functionality, but the more active stereoisomer, 8015-P2, is unique in that it has subnanomolar potency and undergoes entero-hepatic recirculation which extends its in vivo half-life. Daily administration of 8015-P2 to Mfn2 T105M knock-in mice for 6 weeks normalized neuromuscular and sensory dysfunction and corrected histological/ultrastructural neurodegeneration and neurogenic myoatrophy. These studies describe a more clinically relevant mouse model of CMT2A and an improved mitofusin activator derived from piperine. We posit that 8015-P2 and other piperine derivatives may benefit CMT2A or other neurodegenerative conditions wherein mitochondrial dysdynamism plays a contributory role. SIGNIFICANCE STATEMENT: Mitochondrial dysfunction is widespread and broadly contributory in neurodegeneration, but difficult to target therapeutically. Here, we describe 8015-P2, a new small molecule mitofusin activator with ∼10-fold greater potency and improved in vivo pharmacokinetics versus comparators, and demonstrate its rapid reversal of sensory and motor neuron dysfunction in an Mfn2 T105M knock-in mouse model of Charcot-Marie-Tooth disease type 2 A. These findings further support the therapeutic approach of targeting mitochondrial dysdynamism in neurodegeneration.
Collapse
Affiliation(s)
- Jochen Weigele
- Department of Internal Medicine (Pharmacogenomics), Washington University School of Medicine (J.W., L.Z., A.F., E.C., G.W.D.) and Mitochondria in Motion, Inc. (J.W., L.Z.), St. Louis Missouri
| | - Lihong Zhang
- Department of Internal Medicine (Pharmacogenomics), Washington University School of Medicine (J.W., L.Z., A.F., E.C., G.W.D.) and Mitochondria in Motion, Inc. (J.W., L.Z.), St. Louis Missouri
| | - Antonietta Franco
- Department of Internal Medicine (Pharmacogenomics), Washington University School of Medicine (J.W., L.Z., A.F., E.C., G.W.D.) and Mitochondria in Motion, Inc. (J.W., L.Z.), St. Louis Missouri
| | - Etienne Cartier
- Department of Internal Medicine (Pharmacogenomics), Washington University School of Medicine (J.W., L.Z., A.F., E.C., G.W.D.) and Mitochondria in Motion, Inc. (J.W., L.Z.), St. Louis Missouri
| | - Gerald W Dorn
- Department of Internal Medicine (Pharmacogenomics), Washington University School of Medicine (J.W., L.Z., A.F., E.C., G.W.D.) and Mitochondria in Motion, Inc. (J.W., L.Z.), St. Louis Missouri
| |
Collapse
|
9
|
Fu T, Mao C, Chen Z, Huang Y, Li H, Wang C, Liu J, Li S, Lin F. Disease characteristics and clinical specific survival prediction of spinal ependymoma: a genetic and population-based study. Front Neurol 2024; 15:1454061. [PMID: 39346772 PMCID: PMC11428185 DOI: 10.3389/fneur.2024.1454061] [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: 06/24/2024] [Accepted: 07/26/2024] [Indexed: 10/01/2024] Open
Abstract
Background Spinal Ependymoma (SP-EP) is the most commonly occurring tumor affecting the spinal cord. Prompt diagnosis and treatment can significantly enhance prognostic outcomes for patients. In this study, we conducted a comprehensive analysis of RNA sequencing data, along with associated clinical information, from patients diagnosed with SP-EP. The aim was to identify key genes that are characteristic of the disease and develop a survival-related nomogram. Methods We first accessed the Gene Expression Integrated Database (GEO) to acquire the microarray dataset pertaining to SP-EP. This dataset was then processed to identify differentially expressed genes (DEGs) between SP-EP samples and normal controls. Furthermore, machine learning techniques and the CIBERSORT algorithm were employed to extract immune characteristic genes specific to SP-EP patients, thereby enhancing the characterization of target genes. Next, we retrieved comprehensive information on patients diagnosed with SP-EP between 2000 and 2020 from the Surveillance, Epidemiology, and End Results Database (SEER). Using this data, we screened for predictive factors that have a significant impact on patient outcomes. A nomogram was constructed to visualize the predicted overall survival (OS) rates of these patients at 3, 5, and 8 years post-diagnosis. Finally, to assess the reliability and clinical utility of our predictive model, we evaluated it using various metrics including the consistency index (C-index), time-dependent receiver operating characteristic (ROC) curves, area under the curve (AUC), calibration curves, and decision curve analysis (DCA). Results A total of 5,151 DEGs were identified between the SP-EP sample and the normal sample. Analysis of Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways revealed that these DEGs were primarily involved in cellular processes, including cell cycle regulation and cell sensitivity mechanisms. Furthermore, immune infiltration analysis was utilized to identify the core gene CELF4. Regarding the survival rates of patients with SP-EP, the 3-year, 5-year, and 8-year survival rates were 72.5, 57.0, and 40.8%, respectively. Diagnostic age (p < 0.001), gender (p < 0.001), and surgical approach (p < 0.005) were identified as independent prognostic factors for OS. Additionally, a nomogram model was constructed based on these prognostic factors, demonstrating good consistency between predicted and actual results in the study's validation process. Notably, the study also demonstrated that more extensive surgical resection could extend patients' OS. Conclusion Through bioinformatics analysis of microarray datasets, we identified CELF4 as a central gene associated with immune infiltration among DEGs. Previous studies have demonstrated that CELF4 may play a pivotal role in the pathogenesis of SP-EP. Furthermore, this study developed and validated a prognostic prediction model in the form of a nomogram utilizing the SEER database, enabling clinicians to accurately assess treatment risks and benefits, thereby enhancing personalized therapeutic strategies and prognosis predictions.
Collapse
Affiliation(s)
- Tengyue Fu
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
- The Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Chuxiao Mao
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Zhuming Chen
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Yuxiang Huang
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Houlin Li
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Chunhua Wang
- College of Life Science and Technology, Mudanjiang Normal University, Mudanjiang, China
| | - Jie Liu
- The Department of Neurosurgery, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Shenyu Li
- The Department of Neurosurgery, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Famu Lin
- The Department of Neurosurgery, Shunde Hospital of Southern Medical University, Foshan, China
| |
Collapse
|
10
|
Papadimitriou E, Thomaidou D. Post-transcriptional mechanisms controlling neurogenesis and direct neuronal reprogramming. Neural Regen Res 2024; 19:1929-1939. [PMID: 38227517 DOI: 10.4103/1673-5374.390976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 11/08/2023] [Indexed: 01/17/2024] Open
Abstract
Neurogenesis is a tightly regulated process in time and space both in the developing embryo and in adult neurogenic niches. A drastic change in the transcriptome and proteome of radial glial cells or neural stem cells towards the neuronal state is achieved due to sophisticated mechanisms of epigenetic, transcriptional, and post-transcriptional regulation. Understanding these neurogenic mechanisms is of major importance, not only for shedding light on very complex and crucial developmental processes, but also for the identification of putative reprogramming factors, that harbor hierarchically central regulatory roles in the course of neurogenesis and bare thus the capacity to drive direct reprogramming towards the neuronal fate. The major transcriptional programs that orchestrate the neurogenic process have been the focus of research for many years and key neurogenic transcription factors, as well as repressor complexes, have been identified and employed in direct reprogramming protocols to convert non-neuronal cells, into functional neurons. The post-transcriptional regulation of gene expression during nervous system development has emerged as another important and intricate regulatory layer, strongly contributing to the complexity of the mechanisms controlling neurogenesis and neuronal function. In particular, recent advances are highlighting the importance of specific RNA binding proteins that control major steps of mRNA life cycle during neurogenesis, such as alternative splicing, polyadenylation, stability, and translation. Apart from the RNA binding proteins, microRNAs, a class of small non-coding RNAs that block the translation of their target mRNAs, have also been shown to play crucial roles in all the stages of the neurogenic process, from neural stem/progenitor cell proliferation, neuronal differentiation and migration, to functional maturation. Here, we provide an overview of the most prominent post-transcriptional mechanisms mediated by RNA binding proteins and microRNAs during the neurogenic process, giving particular emphasis on the interplay of specific RNA binding proteins with neurogenic microRNAs. Taking under consideration that the molecular mechanisms of neurogenesis exert high similarity to the ones driving direct neuronal reprogramming, we also discuss the current advances in in vitro and in vivo direct neuronal reprogramming approaches that have employed microRNAs or RNA binding proteins as reprogramming factors, highlighting the so far known mechanisms of their reprogramming action.
Collapse
|
11
|
Liu S, Xu X, Omari-Siaw E, Yu J, Deng W. Progress of reprogramming astrocytes into neuron. Mol Cell Neurosci 2024; 130:103947. [PMID: 38862082 DOI: 10.1016/j.mcn.2024.103947] [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: 03/27/2024] [Revised: 06/07/2024] [Accepted: 06/07/2024] [Indexed: 06/13/2024] Open
Abstract
As the main players in the central nervous system (CNS), neurons dominate most life activities. However, after accidental trauma or neurodegenerative diseases, neurons are unable to regenerate themselves. The loss of this important role can seriously affect the quality of life of patients, ranging from movement disorders to disability and even death. There is no suitable treatment to prevent or reverse this process. Therefore, the regeneration of neurons after loss has been a major clinical problem and the key to treatment. Replacing the lost neurons by transdifferentiation of other cells is the only viable approach. Although much progress has been made in stem cell therapy, ethical issues, immune rejection, and limited cell sources still hinder its clinical application. In recent years, somatic cell reprogramming technology has brought a new dawn. Among them, astrocytes, as endogenously abundant cells homologous to neurons, have good potential and application value for reprogramming into neurons, having been reprogrammed into neurons in vitro and in vivo in a variety of ways.
Collapse
Affiliation(s)
- Sitong Liu
- School of Pharmacy, Jiangsu University, Zhenjiang, China; The International Institute on Natural Products and Stem Cells (iNPS), Zhenjiang, China; Key Lab for Drug Delivery & Tissue Regeneration, Zhenjiang, China; Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, China
| | - Ximing Xu
- School of Pharmacy, Jiangsu University, Zhenjiang, China; The International Institute on Natural Products and Stem Cells (iNPS), Zhenjiang, China; Key Lab for Drug Delivery & Tissue Regeneration, Zhenjiang, China; Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, China
| | - Emmanuel Omari-Siaw
- Department of Pharmaceutical Science, Kumasi Technical University, PO Box 854, Kumasi, Ashanti, Ghana
| | - Jiangnan Yu
- School of Pharmacy, Jiangsu University, Zhenjiang, China; The International Institute on Natural Products and Stem Cells (iNPS), Zhenjiang, China; Key Lab for Drug Delivery & Tissue Regeneration, Zhenjiang, China; Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, China.
| | - Wenwen Deng
- School of Pharmacy, Jiangsu University, Zhenjiang, China; The International Institute on Natural Products and Stem Cells (iNPS), Zhenjiang, China; Key Lab for Drug Delivery & Tissue Regeneration, Zhenjiang, China; Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, China.
| |
Collapse
|
12
|
Sun Z, Kwon JS, Ren Y, Chen S, Walker CK, Lu X, Cates K, Karahan H, Sviben S, Fitzpatrick JAJ, Valdez C, Houlden H, Karch CM, Bateman RJ, Sato C, Mennerick SJ, Diamond MI, Kim J, Tanzi RE, Holtzman DM, Yoo AS. Modeling late-onset Alzheimer's disease neuropathology via direct neuronal reprogramming. Science 2024; 385:adl2992. [PMID: 39088624 PMCID: PMC11787906 DOI: 10.1126/science.adl2992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 05/31/2024] [Indexed: 08/03/2024]
Abstract
Late-onset Alzheimer's disease (LOAD) is the most common form of Alzheimer's disease (AD). However, modeling sporadic LOAD that endogenously captures hallmark neuronal pathologies such as amyloid-β (Aβ) deposition, tau tangles, and neuronal loss remains an unmet need. We demonstrate that neurons generated by microRNA (miRNA)-based direct reprogramming of fibroblasts from individuals affected by autosomal dominant AD (ADAD) and LOAD in a three-dimensional environment effectively recapitulate key neuropathological features of AD. Reprogrammed LOAD neurons exhibit Aβ-dependent neurodegeneration, and treatment with β- or γ-secretase inhibitors before (but not subsequent to) Aβ deposit formation mitigated neuronal death. Moreover inhibiting age-associated retrotransposable elements in LOAD neurons reduced both Aβ deposition and neurodegeneration. Our study underscores the efficacy of modeling late-onset neuropathology of LOAD through high-efficiency miRNA-based neuronal reprogramming.
Collapse
Affiliation(s)
- Zhao Sun
- Department of Developmental Biology, Washington University School of Medicine; St. Louis, MO, 63110, USA
- Center for Regenerative Medicine, Washington University School of Medicine; St. Louis, MO, 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine; St. Louis, MO 63110, USA
| | - Ji-Sun Kwon
- Department of Developmental Biology, Washington University School of Medicine; St. Louis, MO, 63110, USA
- Program in Computational and Systems Biology, Washington University School of Medicine; St. Louis, MO 63110, USA
| | - Yudong Ren
- Department of Developmental Biology, Washington University School of Medicine; St. Louis, MO, 63110, USA
- Program in Developmental, Regenerative, and Stem Cell Biology, Washington University School of Medicine; St. Louis, MO 63110, USA
| | - Shawei Chen
- Department of Developmental Biology, Washington University School of Medicine; St. Louis, MO, 63110, USA
- Center for Regenerative Medicine, Washington University School of Medicine; St. Louis, MO, 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine; St. Louis, MO 63110, USA
| | - Courtney K. Walker
- Department of Developmental Biology, Washington University School of Medicine; St. Louis, MO, 63110, USA
- Center for Regenerative Medicine, Washington University School of Medicine; St. Louis, MO, 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine; St. Louis, MO 63110, USA
| | - Xinguo Lu
- Department of Psychiatry, Washington University School of Medicine; St. Louis, MO 63110, USA
| | - Kitra Cates
- Department of Developmental Biology, Washington University School of Medicine; St. Louis, MO, 63110, USA
- Program in Molecular Genetics and Genomics, Washington University School of Medicine; St. Louis, MO 63110, USA
| | - Hande Karahan
- Stark Neurosciences Research Institute, Indiana University School of Medicine; Indianapolis, IN, 46202, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine; Indianapolis, IN, 46202, USA
| | - Sanja Sviben
- Washington University Center for Cellular Imaging, Washington University School of Medicine; St. Louis, MO, 63110, USA
| | - James A J Fitzpatrick
- Washington University Center for Cellular Imaging, Washington University School of Medicine; St. Louis, MO, 63110, USA
| | - Clarissa Valdez
- Center for Alzheimer’s and Neurodegenerative Diseases, Peter O’Donnell Jr. Brain Institute, UT Southwestern Medical Center; Dallas, TX, 75390, USA
| | - Henry Houlden
- UCL Institute of Neurology; Queen Square, London, WC1N 3BG, UK
| | - Celeste M. Karch
- Department of Psychiatry, Washington University School of Medicine; St. Louis, MO 63110, USA
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine; St. Louis, MO 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine; St. Louis, MO 63110, USA
| | - Randall J. Bateman
- Tracy Family SILQ Center for Neurodegenerative Biology; St. Louis, MO, MO 63110, USA
- Department of Neurology, Washington University School of Medicine; St. Louis, MO 63110, USA
| | - Chihiro Sato
- Tracy Family SILQ Center for Neurodegenerative Biology; St. Louis, MO, MO 63110, USA
- Department of Neurology, Washington University School of Medicine; St. Louis, MO 63110, USA
| | - Steven J. Mennerick
- Department of Psychiatry, Washington University School of Medicine; St. Louis, MO 63110, USA
| | - Marc I. Diamond
- Center for Alzheimer’s and Neurodegenerative Diseases, Peter O’Donnell Jr. Brain Institute, UT Southwestern Medical Center; Dallas, TX, 75390, USA
| | - Jungsu Kim
- Stark Neurosciences Research Institute, Indiana University School of Medicine; Indianapolis, IN, 46202, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine; Indianapolis, IN, 46202, USA
| | - Rudolph E. Tanzi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Harvard Medical School; Charlestown, Massachusetts, 02129, USA
| | - David M. Holtzman
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine; St. Louis, MO 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine; St. Louis, MO 63110, USA
- Department of Neurology, Washington University School of Medicine; St. Louis, MO 63110, USA
| | - Andrew S. Yoo
- Department of Developmental Biology, Washington University School of Medicine; St. Louis, MO, 63110, USA
- Center for Regenerative Medicine, Washington University School of Medicine; St. Louis, MO, 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine; St. Louis, MO 63110, USA
| |
Collapse
|
13
|
Nguyen LD, Sengupta S, Cho K, Floru A, George RE, Krichevsky AM. Novel miRNA-inducing drugs enable differentiation of retinoic acid-resistant neuroblastoma cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.05.597584. [PMID: 38895399 PMCID: PMC11185630 DOI: 10.1101/2024.06.05.597584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Tumor cell heterogeneity in neuroblastoma, a pediatric cancer arising from neural crest-derived progenitor cells, poses a significant clinical challenge. In particular, unlike adrenergic (ADRN) neuroblastoma cells, mesenchymal (MES) cells are resistant to chemotherapy and retinoid therapy and thereby significantly contribute to relapses and treatment failures. Previous research suggested that overexpression or activation of miR-124, a neurogenic microRNA with tumor suppressor activity, can induce the differentiation of retinoic acid-resistant neuroblastoma cells. Leveraging our established screen for miRNA-modulatory small molecules, we validated PP121, a dual inhibitor of tyrosine and phosphoinositide kinases, as a robust inducer of miR-124. A combination of PP121 and BDNF-activating bufalin synergistically arrests proliferation, induces differentiation, and maintains the differentiated state of MES SK-N-AS cells for 8 weeks. RNA-seq and deconvolution analyses revealed a collapse of the ADRN core regulatory circuitry (CRC) and the emergence of novel CRCs associated with chromaffin cells and Schwann cell precursors. Using a similar protocol, we differentiated and maintained MES neuroblastoma GI-ME-N and SH-EP cell lines, as well as glioblastoma LN-229 and U-251 cell lines, for over 16 weeks. In conclusion, our novel protocol suggests a promising treatment for therapy-resistant cancers of the nervous system. Moreover, these long-lived, differentiated cells provide valuable models for studying mechanisms underlying differentiation, maturation, and senescence.
Collapse
|
14
|
Mseis-Jackson N, Sharma M, Li H. Controlling the Expression Level of the Neuronal Reprogramming Factors for a Successful Reprogramming Outcome. Cells 2024; 13:1223. [PMID: 39056804 PMCID: PMC11274869 DOI: 10.3390/cells13141223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024] Open
Abstract
Neuronal reprogramming is a promising approach for making major advancement in regenerative medicine. Distinct from the approach of induced pluripotent stem cells, neuronal reprogramming converts non-neuronal cells to neurons without going through a primitive stem cell stage. In vivo neuronal reprogramming brings this approach to a higher level by changing the cell fate of glial cells to neurons in neural tissue through overexpressing reprogramming factors. Despite the ongoing debate over the validation and interpretation of newly generated neurons, in vivo neuronal reprogramming is still a feasible approach and has the potential to become clinical treatment with further optimization and refinement. Here, we discuss the major neuronal reprogramming factors (mostly pro-neurogenic transcription factors during development), especially the significance of their expression levels during neurogenesis and the reprogramming process focusing on NeuroD1. In the developing central nervous system, these pro-neurogenic transcription factors usually elicit distinct spatiotemporal expression patterns that are critical to their function in generating mature neurons. We argue that these dynamic expression patterns may be similarly needed in the process of reprogramming adult cells into neurons and further into mature neurons with subtype identities. We also summarize the existing approaches and propose new ones that control gene expression levels for a successful reprogramming outcome.
Collapse
Affiliation(s)
- Natalie Mseis-Jackson
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
| | - Mehek Sharma
- Department of Biological Sciences, College of Science & Mathematics, Augusta University, Augusta, GA 30912, USA;
| | - Hedong Li
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
| |
Collapse
|
15
|
Tan Z, Qin S, Liu H, Huang X, Pu Y, He C, Yuan Y, Su Z. Small molecules reprogram reactive astrocytes into neuronal cells in the injured adult spinal cord. J Adv Res 2024; 59:111-127. [PMID: 37380102 PMCID: PMC11081968 DOI: 10.1016/j.jare.2023.06.013] [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: 01/24/2023] [Revised: 06/23/2023] [Accepted: 06/25/2023] [Indexed: 06/30/2023] Open
Abstract
INTRODUCTION Ectopic expression of transcription factor-mediated in vivo neuronal reprogramming provides promising strategy to compensate for neuronal loss, while its further clinical application may be hindered by delivery and safety concerns. As a novel and attractive alternative, small molecules may offer a non-viral and non-integrative chemical approach for reprogramming cell fates. Recent definitive evidences have shown that small molecules can convert non-neuronal cells into neurons in vitro. However, whether small molecules alone can induce neuronal reprogramming in vivo remains largely unknown. OBJECTIVES To identify chemical compounds that can induce in vivo neuronal reprogramming in the adult spinal cord. METHODS Immunocytochemistry, immunohistochemistry, qRT-PCR and fate-mapping are performed to analyze the role of small molecules in reprogramming astrocytes into neuronal cells in vitro and in vivo. RESULTS By screening, we identify a chemical cocktail with only two chemical compounds that can directly and rapidly reprogram cultured astrocytes into neuronal cells. Importantly, this chemical cocktail can also successfully trigger neuronal reprogramming in the injured adult spinal cord without introducing exogenous genetic factors. These chemically induced cells showed typical neuronal morphologies and neuron-specific marker expression and could become mature and survive for more than 12 months. Lineage tracing indicated that the chemical compound-converted neuronal cells mainly originated from post-injury spinal reactive astrocytes. CONCLUSION Our proof-of-principle study demonstrates that in vivo glia-to-neuron conversion can be manipulated in a chemical compound-based manner. Albeit our current chemical cocktail has a lowreprogramming efficiency, it will bring in vivo cell fate reprogramming closer to clinical application in brain and spinal cord repair. Future studies should focus on further refining our chemical cocktail and reprogramming approach to boost the reprogramming efficiency.
Collapse
Affiliation(s)
- Zijian Tan
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai 200433, China
| | - Shangyao Qin
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai 200433, China
| | - Hong Liu
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai 200433, China
| | - Xiao Huang
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai 200433, China
| | - Yingyan Pu
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai 200433, China
| | - Cheng He
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai 200433, China
| | - Yimin Yuan
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai 200433, China.
| | - Zhida Su
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
16
|
Aryal S, Chen S, Burbach KF, Yang Y, Capano LS, Kim WK, Bragg DC, Yoo A. SAK3 confers neuroprotection in the neurodegeneration model of X-linked Dystonia-Parkinsonism. RESEARCH SQUARE 2024:rs.3.rs-4068432. [PMID: 38746402 PMCID: PMC11092809 DOI: 10.21203/rs.3.rs-4068432/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Background X-linked Dystonia-Parkinsonism(XDP) is an adult-onset neurodegenerative disorder that results in the loss of striatal medium spiny neurons (MSNs). XDP is associated with disease-specific mutations in and around the TAF1 gene. This study highlights the utility of directly reprogrammed MSNs from fibroblasts of affected XDP individuals as a platform that captures cellular and epigenetic phenotypes associated with XDP-related neurodegeneration. In addition, the current study demonstrates the neuroprotective effect of SAK3 currently tested in other neurodegenerative diseases. Methods XDP fibroblasts from three independent patients as well as age- and sex-matched control fibroblasts were used to generate MSNs by direct neuronal reprogramming using miRNA-9/9*-124 and thetranscription factors CTIP2 , DLX1 -P2A- DLX2 , and MYT1L . Neuronal death, DNA damage, and mitochondrial health assays were carried out to assess the neurodegenerative state of directly reprogrammed MSNs from XDP patients (XDP-MSNs). RNA sequencing and ATAC sequencing were performed to infer changes in the transcriptomic and chromatin landscapesof XDP-MSNs compared to those of control MSNs (Ctrl-MSNs). Results Our results show that XDP patient fibroblasts can be successfully reprogrammed into MSNs and XDP-MSNs display several degenerative phenotypes, including neuronal death, DNA damage, and mitochondrial dysfunction, compared to Ctrl-MSNs reprogrammed from age- and sex-matched control individuals' fibroblasts. In addition, XDP-MSNs showed increased vulnerability to TNFα -toxicity compared to Ctrl-MSNs. To dissect the altered cellular state in XDP-MSNs, we conducted transcriptomic and chromatin accessibility analyses using RNA- and ATAC-seq. Our results indicate that pathways related to neuronal function, calcium signaling, and genes related to other neurodegenerative diseases are commonly altered in XDP-MSNs from multiple patients. Interestingly, we found that SAK3, a T-type calcium channel activator, that may have therapeutic values in other neurodegenerative disorders, protected XDP-MSNs from neuronal death. Notably, we found that SAK3-mediated alleviation of neurodegeneration in XDP-MSNs was accompanied by gene expression changes toward Ctrl-MSNs.
Collapse
|
17
|
Oh YM, Lee SW. Patient-derived neuron model: Capturing age-dependent adult-onset degenerative pathology in Huntington's disease. Mol Cells 2024; 47:100046. [PMID: 38492889 PMCID: PMC11021366 DOI: 10.1016/j.mocell.2024.100046] [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: 12/15/2023] [Revised: 02/27/2024] [Accepted: 03/08/2024] [Indexed: 03/18/2024] Open
Abstract
MicroRNAs play a crucial role in directly reprogramming (converting) human fibroblasts into neurons. Specifically, miR-9/9* and miR-124 (miR-9/9*-124) display neurogenic and cell fate-switching activities when ectopically expressed in human fibroblasts by erasing fibroblast identity and inducing a pan-neuronal state. These converted neurons maintain the biological age of the starting fibroblasts and thus provide a human neuron-based platform to study cellular properties in aged neurons and model adult-onset neurodegenerative disorders using patient-derived cells. Furthermore, the expression of striatal-enriched transcription factors in conjunction with miR-9/9*-124 guides the identity of medium spiny neurons (MSNs), the primary targets in Huntington's disease (HD). Converted MSNs from HD patient-derived fibroblasts (HD-MSNs) can replicate HD-related phenotypes including neurodegeneration associated with age-related declines in critical cellular functions such as autophagy. Here, we review the role of microRNAs in the direct conversion of patient-derived fibroblasts into MSNs and the practical application of converted HD-MSNs as a model for studying adult-onset neuropathology in HD. We provide valuable insights into age-related, cell-intrinsic changes contributing to neurodegeneration in HD-MSNs. Ultimately, we address a comprehensive understanding of the complex molecular landscape underlying HD pathology, offering potential avenues for therapeutic application.
Collapse
Affiliation(s)
- Young Mi Oh
- Department of Biomedical Sciences, Mercer University School of Medicine, Columbus, GA 31901, USA
| | - Seong Won Lee
- Department of Biomedical Sciences, Mercer University School of Medicine, Columbus, GA 31901, USA
| |
Collapse
|
18
|
Burbach KF, Yoo AS. Notch Inhibition Enhances Morphological Reprogramming of microRNA-Induced Human Neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.12.575384. [PMID: 38260259 PMCID: PMC10802628 DOI: 10.1101/2024.01.12.575384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Although the importance of Notch signaling in brain development is well-known, its specific contribution to cellular reprogramming remains less defined. Here, we use microRNA-induced neurons that are directly reprogrammed from human fibroblasts to determine how Notch signaling contributes to neuronal identity. We found that inhibiting Notch signaling led to an increase in neurite extension, while activating Notch signaling had the opposite effect. Surprisingly, Notch inhibition during the first week of reprogramming was both necessary and sufficient to enhance neurite outgrowth at a later timepoint. This timeframe is when the reprogramming miRNAs, miR-9/9* and miR-124, primarily induce a post-mitotic state and erase fibroblast identity. Accordingly, transcriptomic analysis showed that the effect of Notch inhibition was likely due to improvements in fibroblast fate erasure and silencing of anti-neuronal genes. To this effect, we identify MYLIP , whose downregulation in response to Notch inhibition significantly promoted neurite outgrowth. Moreover, Notch inhibition resulted in cells with neuronal transcriptome signature defined by expressing long genes at a faster rate than the control, demonstrating the effect of accelerated fate erasure on neuronal fate acquisition. Our results demonstrate the critical role of Notch signaling in mediating morphological changes in miRNA-based neuronal reprogramming of human adult fibroblasts.
Collapse
|
19
|
Lee SW, Oh YM, Victor MB, Yang Y, Chen S, Strunilin I, Dahiya S, Dolle RE, Pak SC, Silverman GA, Perlmutter DH, Yoo AS. Longitudinal modeling of human neuronal aging reveals the contribution of the RCAN1-TFEB pathway to Huntington's disease neurodegeneration. NATURE AGING 2024; 4:95-109. [PMID: 38066314 PMCID: PMC11456361 DOI: 10.1038/s43587-023-00538-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 11/03/2023] [Indexed: 12/19/2023]
Abstract
Aging is a common risk factor in neurodegenerative disorders. Investigating neuronal aging in an isogenic background stands to facilitate analysis of the interplay between neuronal aging and neurodegeneration. Here we perform direct neuronal reprogramming of longitudinally collected human fibroblasts to reveal genetic pathways altered at different ages. Comparative transcriptome analysis of longitudinally aged striatal medium spiny neurons (MSNs) in Huntington's disease identified pathways involving RCAN1, a negative regulator of calcineurin. Notably, RCAN1 protein increased with age in reprogrammed MSNs as well as in human postmortem striatum and RCAN1 knockdown rescued patient-derived MSNs of Huntington's disease from degeneration. RCAN1 knockdown enhanced chromatin accessibility of genes involved in longevity and autophagy, mediated through enhanced calcineurin activity, leading to TFEB's nuclear localization by dephosphorylation. Furthermore, G2-115, an analog of glibenclamide with autophagy-enhancing activities, reduced the RCAN1-calcineurin interaction, phenocopying the effect of RCAN1 knockdown. Our results demonstrate that targeting RCAN1 genetically or pharmacologically can increase neuronal resilience in Huntington's disease.
Collapse
Affiliation(s)
- Seong Won Lee
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Biomedical Sciences, Mercer University School of Medicine, Columbus, GA, USA
| | - Young Mi Oh
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Biomedical Sciences, Mercer University School of Medicine, Columbus, GA, USA
| | - Matheus B Victor
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yan Yang
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Shawei Chen
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Ilya Strunilin
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Sonika Dahiya
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Roland E Dolle
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA
| | - Stephen C Pak
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Gary A Silverman
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - David H Perlmutter
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrew S Yoo
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA.
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
20
|
Pai C, Sengupta R, Heuckeroth RO. Sequencing Reveals miRNAs Enriched in the Developing Mouse Enteric Nervous System. Noncoding RNA 2023; 10:1. [PMID: 38250801 PMCID: PMC10801555 DOI: 10.3390/ncrna10010001] [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: 11/09/2023] [Revised: 12/01/2023] [Accepted: 12/16/2023] [Indexed: 01/23/2024] Open
Abstract
The enteric nervous system (ENS) is an essential network of neurons and glia in the bowel wall. Defects in ENS development can result in Hirschsprung disease (HSCR), a life-threatening condition characterized by severe constipation, abdominal distention, bilious vomiting, and failure to thrive. A growing body of literature connects HSCR to alterations in miRNA expression, but there are limited data on the normal miRNA landscape in the developing ENS. We sequenced small RNAs (smRNA-seq) and messenger RNAs (mRNA-seq) from ENS precursor cells of mid-gestation Ednrb-EGFP mice and compared them to aggregated RNA from all other cells in the developing bowel. Our smRNA-seq results identified 73 miRNAs that were significantly enriched and highly expressed in the developing ENS, with miR-9, miR-27b, miR-124, miR-137, and miR-488 as our top 5 miRNAs that are conserved in humans. However, contrary to prior reports, our follow-up analyses of miR-137 showed that loss of Mir137 in Nestin-cre, Wnt1-cre, Sox10-cre, or Baf53b-cre lineage cells had no effect on mouse survival or ENS development. Our data provide important context for future studies of miRNAs in HSCR and other ENS diseases and highlight open questions about facility-specific factors in development.
Collapse
Affiliation(s)
- Christopher Pai
- The Children’s Hospital of Philadelphia Research Institute, Philadelphia, PA 19104, USA;
- Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rajarshi Sengupta
- American Association for Cancer Research, Philadelphia, PA 19106, USA;
| | - Robert O. Heuckeroth
- The Children’s Hospital of Philadelphia Research Institute, Philadelphia, PA 19104, USA;
- Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
21
|
Yang ZZ, Parchem RJ. The role of noncoding RNAs in pancreatic birth defects. Birth Defects Res 2023; 115:1785-1808. [PMID: 37066622 PMCID: PMC10579456 DOI: 10.1002/bdr2.2178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/19/2023] [Accepted: 04/03/2023] [Indexed: 04/18/2023]
Abstract
Congenital defects in the pancreas can cause severe health issues such as pancreatic cancer and diabetes which require lifelong treatment. Regenerating healthy pancreatic cells to replace malfunctioning cells has been considered a promising cure for pancreatic diseases including birth defects. However, such therapies are currently unavailable in the clinic. The developmental gene regulatory network underlying pancreatic development must be reactivated for in vivo regeneration and recapitulated in vitro for cell replacement therapy. Thus, understanding the mechanisms driving pancreatic development will pave the way for regenerative therapies. Pancreatic progenitor cells are the precursors of all pancreatic cells which use epigenetic changes to control gene expression during differentiation to generate all of the distinct pancreatic cell types. Epigenetic changes involving DNA methylation and histone modifications can be controlled by noncoding RNAs (ncRNAs). Indeed, increasing evidence suggests that ncRNAs are indispensable for proper organogenesis. Here, we summarize recent insight into the role of ncRNAs in the epigenetic regulation of pancreatic development. We further discuss how disruptions in ncRNA biogenesis and expression lead to developmental defects and diseases. This review summarizes in vivo data from animal models and in vitro studies using stem cell differentiation as a model for pancreatic development.
Collapse
Affiliation(s)
- Ziyue Zoey Yang
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, Texas, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Ronald J Parchem
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, Texas, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
22
|
Zhu B, Fisher E, Li L, Zhong P, Yan Z, Feng J. PTBP2 attenuation facilitates fibroblast to neuron conversion by promoting alternative splicing of neuronal genes. Stem Cell Reports 2023; 18:2268-2282. [PMID: 37832540 PMCID: PMC10679656 DOI: 10.1016/j.stemcr.2023.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 09/19/2023] [Accepted: 09/20/2023] [Indexed: 10/15/2023] Open
Abstract
The direct conversion of human skin fibroblasts to neurons has a low efficiency and unclear mechanism. Here, we show that the knockdown of PTBP2 significantly enhanced the transdifferentiation induced by ASCL1, MIR9/9∗-124, and p53 shRNA (AMp) to generate mostly GABAergic neurons. Longitudinal RNA sequencing analyses identified the continuous induction of many RNA splicing regulators. Among these, the knockdown of RBFOX3 (NeuN), significantly abrogated the transdifferentiation. Overexpression of RBFOX3 significantly enhanced the conversion induced by AMp; the enhancement was occluded by PTBP2 knockdown. We found that PTBP2 attenuation significantly favored neuron-specific alternative splicing (AS) of many genes involved in synaptic transmission, signal transduction, and axon formation. RBFOX3 knockdown significantly reversed the effect, while RBFOX3 overexpression occluded the enhancement. The study reveals the critical role of neuron-specific AS in the direct conversion of human skin fibroblasts to neurons by showing that PTBP2 attenuation enhances this mechanism in concert with RBFOX3.
Collapse
Affiliation(s)
- Binglin Zhu
- Veterans Affairs Western New York Healthcare System, Buffalo, NY 14215, USA; Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Emily Fisher
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Li Li
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Ping Zhong
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Zhen Yan
- Veterans Affairs Western New York Healthcare System, Buffalo, NY 14215, USA; Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Jian Feng
- Veterans Affairs Western New York Healthcare System, Buffalo, NY 14215, USA; Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY 14203, USA.
| |
Collapse
|
23
|
Fang YM, Chen WC, Zheng WJ, Yang YS, Zhang Y, Chen XL, Pei MQ, Lin S, He HF. A cutting-edge strategy for spinal cord injury treatment: resident cellular transdifferentiation. Front Cell Neurosci 2023; 17:1237641. [PMID: 37711511 PMCID: PMC10498389 DOI: 10.3389/fncel.2023.1237641] [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: 06/09/2023] [Accepted: 08/14/2023] [Indexed: 09/16/2023] Open
Abstract
Spinal cord injury causes varying degrees of motor and sensory function loss. However, there are no effective treatments for spinal cord repair following an injury. Moreover, significant preclinical advances in bioengineering and regenerative medicine have not yet been translated into effective clinical therapies. The spinal cord's poor regenerative capacity makes repairing damaged and lost neurons a critical treatment step. Reprogramming-based neuronal transdifferentiation has recently shown great potential in repair and plasticity, as it can convert mature somatic cells into functional neurons for spinal cord injury repair in vitro and in vivo, effectively halting the progression of spinal cord injury and promoting functional improvement. However, the mechanisms of the neuronal transdifferentiation and the induced neuronal subtypes are not yet well understood. This review analyzes the mechanisms of resident cellular transdifferentiation based on a review of the relevant recent literature, describes different molecular approaches to obtain different neuronal subtypes, discusses the current challenges and improvement methods, and provides new ideas for exploring therapeutic approaches for spinal cord injury.
Collapse
Affiliation(s)
- Yu-Ming Fang
- Department of Anaesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Wei-Can Chen
- Department of Anaesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Wan-Jing Zheng
- Department of Anaesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Yu-Shen Yang
- Department of Anaesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Yan Zhang
- Department of Anaesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Xin-Li Chen
- Department of Anaesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Meng-Qin Pei
- Department of Anaesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Shu Lin
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- Neuroendocrinology Group, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - He-Fan He
- Department of Anaesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| |
Collapse
|
24
|
Steadman K, You S, Srinivas DV, Mouakkad L, Yan Y, Kim M, Venugopal SV, Tanaka H, Freeman MR. Autonomous action and cooperativity between the ONECUT2 transcription factor and its 3' untranslated region. Front Cell Dev Biol 2023; 11:1206259. [PMID: 37484909 PMCID: PMC10356556 DOI: 10.3389/fcell.2023.1206259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 05/16/2023] [Indexed: 07/25/2023] Open
Abstract
The transcription factor ONECUT2 (OC2) is a master transcriptional regulator operating in metastatic castration-resistant prostate cancer that suppresses androgen receptor activity and promotes neural differentiation and tumor cell survival. OC2 mRNA possesses an unusually long (14,575 nt), evolutionarily conserved 3' untranslated region (3' UTR) with many microRNA binding sites, including up to 26 miR-9 sites. This is notable because miR-9 targets many of the same genes regulated by the OC2 protein. Paradoxically, OC2 expression is high in tissues with high miR-9 expression. The length and complex secondary structure of OC2 mRNA suggests that it is a potent master competing endogenous RNA (ceRNA) capable of sequestering miRNAs. Here, we describe a novel role for OC2 3' UTR in lethal prostate cancer consistent with a function as a ceRNA. A plausible ceRNA network in OC2-driven tumors was constructed computationally and then confirmed in prostate cancer cell lines. Genes regulated by OC2 3' UTR exhibited high overlap (up to 45%) with genes driven by the overexpression of the OC2 protein in the absence of 3' UTR, indicating a cooperative functional relationship between the OC2 protein and its 3' UTR. These overlapping networks suggest an evolutionarily conserved mechanism to reinforce OC2 transcription by protection of OC2-regulated mRNAs from miRNA suppression. Both the protein and 3' UTR showed increased polycomb-repressive complex activity. The expression of OC2 3' UTR mRNA alone (without protein) dramatically increased the metastatic potential by in vitro assays. Additionally, OC2 3' UTR increased the expression of Aldo-Keto reductase and UDP-glucuronyl transferase family genes responsible for altering the androgen synthesis pathway. ONECUT2 represents the first-described dual-modality transcript that operates as both a key transcription factor driving castration-resistant prostate cancer and a master ceRNA that promotes and protects the same transcriptional network.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Michael R. Freeman
- Division of Cancer Biology and Therapeutics, Biomedical Sciences and Pathology and Laboratory Medicine, Department of Urology, Cedars-Sinai Medical Center, Samuel Oschin Comprehensive Cancer Institute, Los Angeles, CA, United States
| |
Collapse
|
25
|
Lear BP, Moore DL. Moving CNS axon growth and regeneration research into human model systems. Front Neurosci 2023; 17:1198041. [PMID: 37425013 PMCID: PMC10324669 DOI: 10.3389/fnins.2023.1198041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/25/2023] [Indexed: 07/11/2023] Open
Abstract
Axon regeneration is limited in the adult mammalian central nervous system (CNS) due to both intrinsic and extrinsic factors. Rodent studies have shown that developmental age can drive differences in intrinsic axon growth ability, such that embryonic rodent CNS neurons extend long axons while postnatal and adult CNS neurons do not. In recent decades, scientists have identified several intrinsic developmental regulators in rodents that modulate growth. However, whether this developmentally programmed decline in CNS axon growth is conserved in humans is not yet known. Until recently, there have been limited human neuronal model systems, and even fewer age-specific human models. Human in vitro models range from pluripotent stem cell-derived neurons to directly reprogrammed (transdifferentiated) neurons derived from human somatic cells. In this review, we discuss the advantages and disadvantages of each system, and how studying axon growth in human neurons can provide species-specific knowledge in the field of CNS axon regeneration with the goal of bridging basic science studies to clinical trials. Additionally, with the increased availability and quality of 'omics datasets of human cortical tissue across development and lifespan, scientists can mine these datasets for developmentally regulated pathways and genes. As there has been little research performed in human neurons to study modulators of axon growth, here we provide a summary of approaches to begin to shift the field of CNS axon growth and regeneration into human model systems to uncover novel drivers of axon growth.
Collapse
Affiliation(s)
| | - Darcie L. Moore
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
26
|
Sun Z, Kwon JS, Ren Y, Chen S, Cates K, Lu X, Walker CK, Karahan H, Sviben S, Fitzpatrick JAJ, Valdez C, Houlden H, Karch CM, Bateman RJ, Sato C, Mennerick SJ, Diamond MI, Kim J, Tanzi RE, Holtzman DM, Yoo AS. Endogenous recapitulation of Alzheimer's disease neuropathology through human 3D direct neuronal reprogramming. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.24.542155. [PMID: 37292658 PMCID: PMC10245934 DOI: 10.1101/2023.05.24.542155] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that primarily affects elderly individuals, and is characterized by hallmark neuronal pathologies including extracellular amyloid-β (Aβ) plaque deposition, intracellular tau tangles, and neuronal death. However, recapitulating these age-associated neuronal pathologies in patient-derived neurons has remained a significant challenge, especially for late-onset AD (LOAD), the most common form of the disorder. Here, we applied the high efficiency microRNA-mediated direct neuronal reprogramming of fibroblasts from AD patients to generate cortical neurons in three-dimensional (3D) Matrigel and self-assembled neuronal spheroids. Our findings indicate that neurons and spheroids reprogrammed from both autosomal dominant AD (ADAD) and LOAD patients exhibited AD-like phenotypes linked to neurons, including extracellular Aβ deposition, dystrophic neurites with hyperphosphorylated, K63-ubiquitin-positive, seed-competent tau, and spontaneous neuronal death in culture. Moreover, treatment with β- or γ-secretase inhibitors in LOAD patient-derived neurons and spheroids before Aβ deposit formation significantly lowered Aβ deposition, as well as tauopathy and neurodegeneration. However, the same treatment after the cells already formed Aβ deposits only had a mild effect. Additionally, inhibiting the synthesis of age-associated retrotransposable elements (RTEs) by treating LOAD neurons and spheroids with the reverse transcriptase inhibitor, lamivudine, alleviated AD neuropathology. Overall, our results demonstrate that direct neuronal reprogramming of AD patient fibroblasts in a 3D environment can capture age-related neuropathology and reflect the interplay between Aβ accumulation, tau dysregulation, and neuronal death. Moreover, miRNA-based 3D neuronal conversion provides a human-relevant AD model that can be used to identify compounds that can potentially ameliorate AD-associated pathologies and neurodegeneration.
Collapse
|
27
|
Wang B, Li C, Ming J, Wu L, Fang S, Huang Y, Lin L, Liu H, Kuang J, Zhao C, Huang X, Feng H, Guo J, Yang X, Guo L, Zhang X, Chen J, Liu J, Zhu P, Pei D. The NuRD complex cooperates with SALL4 to orchestrate reprogramming. Nat Commun 2023; 14:2846. [PMID: 37208322 DOI: 10.1038/s41467-023-38543-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 05/08/2023] [Indexed: 05/21/2023] Open
Abstract
Cell fate decision involves rewiring of the genome, but remains poorly understood at the chromatin level. Here, we report that chromatin remodeling complex NuRD participates in closing open chromatin in the early phase of somatic reprogramming. Sall4, Jdp2, Glis1 and Esrrb can reprogram MEFs to iPSCs efficiently, but only Sall4 is indispensable capable of recruiting endogenous components of NuRD. Yet knocking down NuRD components only reduces reprogramming modestly, in contrast to disrupting the known Sall4-NuRD interaction by mutating or deleting the NuRD interacting motif at its N-terminus that renders Sall4 inept to reprogram. Remarkably, these defects can be partially rescured by grafting NuRD interacting motif onto Jdp2. Further analysis of chromatin accessibility dynamics demonstrates that the Sall4-NuRD axis plays a critical role in closing the open chromatin in the early phase of reprogramming. Among the chromatin loci closed by Sall4-NuRD encode genes resistant to reprogramming. These results identify a previously unrecognized role of NuRD in reprogramming, and may further illuminate chromatin closing as a critical step in cell fate control.
Collapse
Affiliation(s)
- Bo Wang
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China
- Center for Cell Lineage and Atlas (CCLA), Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
- CAS Key Laboratory of Regenerative Biology, South China Institutes for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Chen Li
- CAS Key Laboratory of Regenerative Biology, South China Institutes for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academic of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jin Ming
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China
- CAS Key Laboratory of Regenerative Biology, South China Institutes for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academic of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Linlin Wu
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China
| | - Shicai Fang
- CAS Key Laboratory of Regenerative Biology, South China Institutes for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academic of Sciences, Guangzhou, China
- Joint School of Life Science, Guangzhou Medical University-Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yi Huang
- CAS Key Laboratory of Regenerative Biology, South China Institutes for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academic of Sciences, Guangzhou, China
- Joint School of Life Science, Guangzhou Medical University-Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Lihui Lin
- Center for Cell Lineage and Atlas (CCLA), Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
- CAS Key Laboratory of Regenerative Biology, South China Institutes for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academic of Sciences, Guangzhou, China
- Guangzhou Branch of the Supercomputing Center of Chinese Academy of Sciences, Guangzhou, China
| | - He Liu
- Center for Cell Lineage and Atlas (CCLA), Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
- CAS Key Laboratory of Regenerative Biology, South China Institutes for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academic of Sciences, Guangzhou, China
| | - Junqi Kuang
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China
- CAS Key Laboratory of Regenerative Biology, South China Institutes for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academic of Sciences, Guangzhou, China
- Guangzhou Branch of the Supercomputing Center of Chinese Academy of Sciences, Guangzhou, China
| | - Chengchen Zhao
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China
| | - Xingnan Huang
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China
| | - Huijian Feng
- Center for Cell Lineage and Atlas (CCLA), Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
- CAS Key Laboratory of Regenerative Biology, South China Institutes for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academic of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jing Guo
- CAS Key Laboratory of Regenerative Biology, South China Institutes for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academic of Sciences, Guangzhou, China
| | - Xuejie Yang
- CAS Key Laboratory of Regenerative Biology, South China Institutes for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academic of Sciences, Guangzhou, China
| | - Liman Guo
- Center for Cell Lineage and Atlas (CCLA), Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Xiaofei Zhang
- Center for Cell Lineage and Atlas (CCLA), Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
- CAS Key Laboratory of Regenerative Biology, South China Institutes for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academic of Sciences, Guangzhou, China
| | - Jiekai Chen
- Center for Cell Lineage and Atlas (CCLA), Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
- CAS Key Laboratory of Regenerative Biology, South China Institutes for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academic of Sciences, Guangzhou, China
| | - Jing Liu
- Center for Cell Lineage and Atlas (CCLA), Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
- CAS Key Laboratory of Regenerative Biology, South China Institutes for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academic of Sciences, Guangzhou, China
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, China.
| | - Duanqing Pei
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China.
| |
Collapse
|
28
|
Papadimitriou E, Koutsoudaki PN, Thanou I, Karagkouni D, Karamitros T, Chroni-Tzartou D, Gaitanou M, Gkemisis C, Margariti M, Xingi E, Tzartos SJ, Hatzigeorgiou AG, Thomaidou D. A miR-124-mediated post-transcriptional mechanism controlling the cell fate switch of astrocytes to induced neurons. Stem Cell Reports 2023; 18:915-935. [PMID: 36963393 PMCID: PMC10147664 DOI: 10.1016/j.stemcr.2023.02.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 02/21/2023] [Accepted: 02/23/2023] [Indexed: 03/26/2023] Open
Abstract
The microRNA (miRNA) miR-124 has been employed supplementary to neurogenic transcription factors (TFs) and other miRNAs to enhance direct neurogenic conversion. The aim of this study was to investigate whether miR-124 is sufficient to drive direct reprogramming of astrocytes to induced neurons (iNs) on its own and elucidate its independent mechanism of reprogramming action. Our data show that miR-124 is a potent driver of the reprogramming switch of astrocytes toward an immature neuronal fate by directly targeting the RNA-binding protein Zfp36L1 implicated in ARE-mediated mRNA decay and subsequently derepressing Zfp36L1 neurogenic interactome. To this end, miR-124 contribution in iNs' production largely recapitulates endogenous neurogenesis pathways, being further enhanced upon addition of the neurogenic compound ISX9, which greatly improves iNs' differentiation and functional maturation. Importantly, miR-124 is potent in guiding direct conversion of reactive astrocytes to immature iNs in vivo following cortical trauma, while ISX9 supplementation confers a survival advantage to newly produced iNs.
Collapse
Affiliation(s)
- Elsa Papadimitriou
- Neural Stem Cells and Neuroimaging Group, Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| | - Paraskevi N Koutsoudaki
- Neural Stem Cells and Neuroimaging Group, Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| | - Irini Thanou
- Neural Stem Cells and Neuroimaging Group, Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| | - Dimitra Karagkouni
- DIANA-Lab, Hellenic Pasteur Institute & Department of Computer Science and Biomedical Informatics, University of Thessaly, Larissa, Greece
| | - Timokratis Karamitros
- Bioinformatics and Applied Genomics Unit, Department of Microbiology, Hellenic Pasteur Institute, Athens, Greece
| | - Dafni Chroni-Tzartou
- Laboratory of Molecular Neurobiology and Immunology, Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| | - Maria Gaitanou
- Laboratory of Cellular and Molecular Neurobiology - Stem Cells, Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| | - Christos Gkemisis
- Neural Stem Cells and Neuroimaging Group, Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| | - Maria Margariti
- Neural Stem Cells and Neuroimaging Group, Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| | - Evangelia Xingi
- Light Microscopy Unit, Hellenic Pasteur Institute, Athens, Greece
| | - Socrates J Tzartos
- Laboratory of Molecular Neurobiology and Immunology, Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| | - Artemis G Hatzigeorgiou
- Bioinformatics and Applied Genomics Unit, Department of Microbiology, Hellenic Pasteur Institute, Athens, Greece
| | - Dimitra Thomaidou
- Neural Stem Cells and Neuroimaging Group, Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece; Light Microscopy Unit, Hellenic Pasteur Institute, Athens, Greece.
| |
Collapse
|
29
|
Dang X, Zhang L, Franco A, Dorn II GW. Activating mitofusins interrupts mitochondrial degeneration and delays disease progression in SOD1 mutant amyotrophic lateral sclerosis. Hum Mol Genet 2023; 32:1208-1222. [PMID: 36416308 PMCID: PMC10026224 DOI: 10.1093/hmg/ddac287] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/25/2022] [Accepted: 11/21/2022] [Indexed: 11/24/2022] Open
Abstract
Mitochondrial involvement in neurodegenerative diseases is widespread and multifactorial. Targeting mitochondrial pathology is therefore of interest. The recent development of bioactive molecules that modulate mitochondrial dynamics (fusion, fission and motility) offers a new therapeutic approach for neurodegenerative diseases with either indirect or direct mitochondrial involvement. Here, we asked: (1) Can enhanced mitochondrial fusion and motility improve secondary mitochondrial pathology in superoxide dismutase1 (SOD1) mutant amyotrophic lateral sclerosis (ALS)? And: (2) What is the impact of enhancing mitochondria fitness on in vivo manifestations of SOD1 mutant ALS? We observed that small molecule mitofusin activators corrected mitochondrial fragmentation, depolarization and dysmotility in genetically diverse ALS patient reprogrammed motor neurons and fibroblasts, and in motor neurons, sensory neurons and fibroblasts from SOD1 G93A mice. Continuous, but not intermittent, pharmacologic mitofusin activation delayed phenotype progression and lethality in SOD1 G93A mice, reducing neuron loss and improving neuromuscular connectivity. Mechanistically, mitofusin activation increased mitochondrial motility, fitness and residency within neuromuscular synapses; reduced mitochondrial reactive oxygen species production; and diminished apoptosis in SOD1 mutant neurons. These benefits were accompanied by improved mitochondrial respiratory coupling, despite characteristic SOD1 mutant ALS-associated downregulation of mitochondrial respiratory complexes. Targeting mitochondrial dysdynamism is a promising approach to alleviate pathology caused by secondary mitochondrial dysfunction in some neurodegenerative diseases.
Collapse
Affiliation(s)
- Xiawei Dang
- Department of Internal Medicine, Washington University School of Medicine, St. Louis MO USA
- Department of Psychiatry, The First Affiliated Hospital of Xi’an Jiao Tong University, Xi’an, Shaanxi 710061, China
| | - Lihong Zhang
- Department of Internal Medicine, Washington University School of Medicine, St. Louis MO USA
| | - Antonietta Franco
- Department of Internal Medicine, Washington University School of Medicine, St. Louis MO USA
| | - Gerald W Dorn II
- Department of Internal Medicine, Washington University School of Medicine, St. Louis MO USA
| |
Collapse
|
30
|
Dennison R, Usuga E, Chen H, Paul JZ, Arbelaez CA, Teng YD. Direct Cell Reprogramming and Phenotypic Conversion: An Analysis of Experimental Attempts to Transform Astrocytes into Neurons in Adult Animals. Cells 2023; 12:618. [PMID: 36831283 PMCID: PMC9954435 DOI: 10.3390/cells12040618] [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: 11/29/2022] [Revised: 02/06/2023] [Accepted: 02/09/2023] [Indexed: 02/17/2023] Open
Abstract
Central nervous system (CNS) repair after injury or disease remains an unresolved problem in neurobiology research and an unmet medical need. Directly reprogramming or converting astrocytes to neurons (AtN) in adult animals has been investigated as a potential strategy to facilitate brain and spinal cord recovery and advance fundamental biology. Conceptually, AtN strategies rely on forced expression or repression of lineage-specific transcription factors to make endogenous astrocytes become "induced neurons" (iNs), presumably without re-entering any pluripotent or multipotent states. The AtN-derived cells have been reported to manifest certain neuronal functions in vivo. However, this approach has raised many new questions and alternative explanations regarding the biological features of the end products (e.g., iNs versus neuron-like cells, neural functional changes, etc.), developmental biology underpinnings, and neurobiological essentials. For this paper per se, we proposed to draw an unconventional distinction between direct cell conversion and direct cell reprogramming, relative to somatic nuclear transfer, based on the experimental methods utilized to initiate the transformation process, aiming to promote a more in-depth mechanistic exploration. Moreover, we have summarized the current tactics employed for AtN induction, comparisons between the bench endeavors concerning outcome tangibility, and discussion of the issues of published AtN protocols. Lastly, the urgency to clearly define/devise the theoretical frameworks, cell biological bases, and bench specifics to experimentally validate primary data of AtN studies was highlighted.
Collapse
Affiliation(s)
- Rachel Dennison
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA 02129, USA
- Laboratory of SCI, Stem Cell and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical School, Boston, MA 02115, USA
| | - Esteban Usuga
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA 02129, USA
- Laboratory of SCI, Stem Cell and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical School, Boston, MA 02115, USA
| | - Harriet Chen
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA 02129, USA
- Laboratory of SCI, Stem Cell and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical School, Boston, MA 02115, USA
| | - Jacob Z. Paul
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA 02129, USA
- Laboratory of SCI, Stem Cell and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical School, Boston, MA 02115, USA
| | - Christian A. Arbelaez
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA 02129, USA
- Laboratory of SCI, Stem Cell and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical School, Boston, MA 02115, USA
| | - Yang D. Teng
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA 02129, USA
- Laboratory of SCI, Stem Cell and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical School, Boston, MA 02115, USA
- Neurotrauma Recovery Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
31
|
Keuls RA, Oh YS, Patel I, Parchem RJ. Post-transcriptional regulation in cranial neural crest cells expands developmental potential. Proc Natl Acad Sci U S A 2023; 120:e2212578120. [PMID: 36724256 PMCID: PMC9963983 DOI: 10.1073/pnas.2212578120] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 12/20/2022] [Indexed: 02/03/2023] Open
Abstract
Developmental potential is progressively restricted after germ layer specification during gastrulation. However, cranial neural crest cells challenge this paradigm, as they develop from anterior ectoderm, yet give rise to both ectodermal derivatives of the peripheral nervous system and ectomesenchymal bone and cartilage. How cranial neural crest cells differentiate into multiple lineages is poorly understood. Here, we demonstrate that cranial neural crest cells possess a transient state of increased chromatin accessibility. We profile the spatiotemporal emergence of premigratory neural crest and find evidence of lineage bias toward either a neuronal or ectomesenchymal fate, with each expressing distinct factors from earlier stages of development. We identify the miR-302 miRNA family to be highly expressed in cranial neural crest cells and genetic deletion leads to precocious specification of the ectomesenchymal lineage. Loss of mir-302 results in reduced chromatin accessibility in the neuronal progenitor lineage of neural crest and a reduction in peripheral neuron differentiation. Mechanistically, we find that mir-302 directly targets Sox9 to slow the timing of ectomesenchymal neural crest specification and represses multiple genes involved in chromatin condensation to promote accessibility required for neuronal differentiation. Our findings reveal a posttranscriptional mechanism governed by miRNAs to expand developmental potential of cranial neural crest.
Collapse
Affiliation(s)
- Rachel A. Keuls
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX77030
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX77030
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX77030
- Department of Neuroscience, Baylor College of Medicine, Houston, TX77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX77030
| | - Young Sun Oh
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX77030
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX77030
- Department of Neuroscience, Baylor College of Medicine, Houston, TX77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX77030
| | - Ivanshi Patel
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX77030
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX77030
- Department of Neuroscience, Baylor College of Medicine, Houston, TX77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX77030
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX77030
| | - Ronald J. Parchem
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX77030
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX77030
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX77030
- Department of Neuroscience, Baylor College of Medicine, Houston, TX77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX77030
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX77030
| |
Collapse
|
32
|
Nalavade R, Singh M. Intracellular Compartmentalization: A Key Determinant of MicroRNA Functions. Microrna 2023; 12:114-130. [PMID: 37638608 DOI: 10.2174/2211536612666230330184006] [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: 11/24/2022] [Revised: 12/26/2022] [Accepted: 01/19/2023] [Indexed: 08/29/2023]
Abstract
Being an integral part of the eukaryotic transcriptome, miRNAs are regarded as vital regulators of diverse developmental and physiological processes. Clearly, miRNA activity is kept in check by various regulatory mechanisms that control their biogenesis and decay pathways. With the increasing technical depth of RNA profiling technologies, novel insights have unravelled the spatial diversity exhibited by miRNAs inside a cell. Compartmentalization of miRNAs adds complexity to the regulatory circuits of miRNA expression, thereby providing superior control over the miRNA function. This review provides a bird's eye view of miRNAs expressed in different subcellular locations, thus affecting the gene regulatory pathways therein. Occurrence of miRNAs in diverse intracellular locales also reveals various unconventional roles played by miRNAs in different cellular organelles and expands the scope of miRNA functions beyond their traditionally known repressive activities.
Collapse
Affiliation(s)
- Rohit Nalavade
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | - Mohini Singh
- Department of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Greater Noida, India
| |
Collapse
|
33
|
Li J, Xu X, Liu J, Zhang S, Tan X, Li Z, Zhang J, Wang Z. Decoding microRNAs in autism spectrum disorder. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 30:535-546. [PMID: 36457702 PMCID: PMC9685394 DOI: 10.1016/j.omtn.2022.11.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Autism spectrum disorder (ASD)-a congenital mental disorder accompanied by social dysfunction and stereotyped behaviors-has attracted a great deal of attention worldwide. A combination of genetic and environmental factors may determine the pathogenesis of ASD. Recent research of multiple ASD models indicates that microRNAs (miRNAs) play a central role at the onset and progression of ASD by repressing the translation of key mRNAs in neural development and functions. As such, miRNAs show great potential to serve as biomarkers for ASD diagnosis or prognosis and therapeutic targets for the treatment of ASD. In this review, we discuss the regulatory mechanisms by which miRNAs influence ASD phenotypes through various in vivo and in vitro models, including necropsy specimens, animal models, cellular models, and, in particular, induced pluripotent stem cells derived from patients with ASD. We then discuss the potential of miRNA-based therapeutic strategies for ASD currently being evaluated in preclinical studies.
Collapse
Affiliation(s)
- Jinyu Li
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, Shandong 266071, China
| | - Xiaohui Xu
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, Shandong 266071, China
| | - Jiane Liu
- Department of Reproductive Medicine, the Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, China
| | - Sudan Zhang
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, Shandong 266071, China
| | - Xiaohua Tan
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, Shandong 266071, China
| | - Zhiqiang Li
- The Affiliated Hospital of Qingdao University & the Biomedical Sciences Institute of Qingdao University, Qingdao Branch of SJTU Bio-X Institutes, Qingdao University, Qingdao, Shandong 266003, China
| | - Jian Zhang
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Zheng Wang
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, Shandong 266071, China
- Department of Reproductive Medicine, the Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, China
| |
Collapse
|
34
|
Yang R, Pan J, Wang Y, Xia P, Tai M, Jiang Z, Chen G. Application and prospects of somatic cell reprogramming technology for spinal cord injury treatment. Front Cell Neurosci 2022; 16:1005399. [PMID: 36467604 PMCID: PMC9712200 DOI: 10.3389/fncel.2022.1005399] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 11/02/2022] [Indexed: 08/10/2023] Open
Abstract
Spinal cord injury (SCI) is a serious neurological trauma that is challenging to treat. After SCI, many neurons in the injured area die due to necrosis or apoptosis, and astrocytes, oligodendrocytes, microglia and other non-neuronal cells become dysfunctional, hindering the repair of the injured spinal cord. Corrective surgery and biological, physical and pharmacological therapies are commonly used treatment modalities for SCI; however, no current therapeutic strategies can achieve complete recovery. Somatic cell reprogramming is a promising technology that has gradually become a feasible therapeutic approach for repairing the injured spinal cord. This revolutionary technology can reprogram fibroblasts, astrocytes, NG2 cells and neural progenitor cells into neurons or oligodendrocytes for spinal cord repair. In this review, we provide an overview of the transcription factors, genes, microRNAs (miRNAs), small molecules and combinations of these factors that can mediate somatic cell reprogramming to repair the injured spinal cord. Although many challenges and questions related to this technique remain, we believe that the beneficial effect of somatic cell reprogramming provides new ideas for achieving functional recovery after SCI and a direction for the development of treatments for SCI.
Collapse
Affiliation(s)
- Riyun Yang
- Department of Histology and Embryology, Medical School of Nantong University, Nantong, China
| | - Jingying Pan
- Department of Histology and Embryology, Medical School of Nantong University, Nantong, China
| | - Yankai Wang
- Center for Basic Medical Research, Medical School of Nantong University, Nantong, China
| | - Panhui Xia
- Center for Basic Medical Research, Medical School of Nantong University, Nantong, China
| | - Mingliang Tai
- Center for Basic Medical Research, Medical School of Nantong University, Nantong, China
| | - Zhihao Jiang
- Center for Basic Medical Research, Medical School of Nantong University, Nantong, China
| | - Gang Chen
- Center for Basic Medical Research, Medical School of Nantong University, Nantong, China
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
35
|
Oh YM, Lee SW, Kim WK, Chen S, Church VA, Cates K, Li T, Zhang B, Dolle RE, Dahiya S, Pak SC, Silverman GA, Perlmutter DH, Yoo AS. Age-related Huntington's disease progression modeled in directly reprogrammed patient-derived striatal neurons highlights impaired autophagy. Nat Neurosci 2022; 25:1420-1433. [PMID: 36303071 PMCID: PMC10162007 DOI: 10.1038/s41593-022-01185-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 09/19/2022] [Indexed: 01/13/2023]
Abstract
Huntington's disease (HD) is an inherited neurodegenerative disorder with adult-onset clinical symptoms, but the mechanism by which aging drives the onset of neurodegeneration in patients with HD remains unclear. In this study we examined striatal medium spiny neurons (MSNs) directly reprogrammed from fibroblasts of patients with HD to model the age-dependent onset of pathology. We found that pronounced neuronal death occurred selectively in reprogrammed MSNs from symptomatic patients with HD (HD-MSNs) compared to MSNs derived from younger, pre-symptomatic patients (pre-HD-MSNs) and control MSNs from age-matched healthy individuals. We observed age-associated alterations in chromatin accessibility between HD-MSNs and pre-HD-MSNs and identified miR-29b-3p, whose age-associated upregulation promotes HD-MSN degeneration by impairing autophagic function through human-specific targeting of the STAT3 3' untranslated region. Reducing miR-29b-3p or chemically promoting autophagy increased the resilience of HD-MSNs against neurodegeneration. Our results demonstrate miRNA upregulation with aging in HD as a detrimental process driving MSN degeneration and potential approaches for enhancing autophagy and resilience of HD-MSNs.
Collapse
Affiliation(s)
- Young Mi Oh
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Seong Won Lee
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Woo Kyung Kim
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Shawei Chen
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Victoria A Church
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Kitra Cates
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tiandao Li
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Bo Zhang
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Roland E Dolle
- Department of Biochemistry, Washington University School of Medicine, St. Louis, MO, USA
| | - Sonika Dahiya
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Stephen C Pak
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Gary A Silverman
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - David H Perlmutter
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrew S Yoo
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA.
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
36
|
Inagaki E, Yoshimatsu S, Okano H. Accelerated neuronal aging in vitro ∼melting watch ∼. Front Aging Neurosci 2022; 14:868770. [PMID: 36016855 PMCID: PMC9397486 DOI: 10.3389/fnagi.2022.868770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
In developed countries, the aging of the population and the associated increase in age-related diseases are causing major unresolved medical, social, and environmental matters. Therefore, research on aging has become one of the most important and urgent issues in life sciences. If the molecular mechanisms of the onset and progression of neurodegenerative diseases are elucidated, we can expect to develop disease-modifying methods to prevent neurodegeneration itself. Since the discovery of induced pluripotent stem cells (iPSCs), there has been an explosion of disease models using disease-specific iPSCs derived from patient-derived somatic cells. By inducing the differentiation of iPSCs into neurons, disease models that reflect the patient-derived pathology can be reproduced in culture dishes, and are playing an active role in elucidating new pathological mechanisms and as a platform for new drug discovery. At the same time, however, we are faced with a new problem: how to recapitulate aging in culture dishes. It has been pointed out that cells differentiated from pluripotent stem cells are juvenile, retain embryonic traits, and may not be fully mature. Therefore, attempts are being made to induce cell maturation, senescence, and stress signals through culture conditions. It has also been reported that direct conversion of fibroblasts into neurons can reproduce human neurons with an aged phenotype. Here, we outline some state-of-the-art insights into models of neuronal aging in vitro. New frontiers in which stem cells and methods for inducing differentiation of tissue regeneration can be applied to aging research are just now approaching, and we need to keep a close eye on them. These models are forefront and intended to advance our knowledge of the molecular mechanisms of aging and contribute to the development of novel therapies for human neurodegenerative diseases associated with aging.
Collapse
Affiliation(s)
- Emi Inagaki
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
- Japanese Society for the Promotion of Science (JSPS), Tokyo, Japan
| | - Sho Yoshimatsu
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
- *Correspondence: Hideyuki Okano,
| |
Collapse
|
37
|
Lee S, Shim HS, Park HJ, Chang Y, Han YE, Oh SJ, Lee W, Im H, Seol Y, Ryu H, Kang H, Lee YK, Park S, Yoo J. Elongated nanoporous Au networks improve somatic cell direct conversion into induced dopaminergic neurons for Parkinson's disease therapy. Acta Biomater 2022; 151:561-575. [PMID: 35931279 DOI: 10.1016/j.actbio.2022.07.058] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 07/26/2022] [Accepted: 07/28/2022] [Indexed: 11/18/2022]
Abstract
The efficient production of dopaminergic neurons via the direct conversion of other cell types is of interest as a potential therapeutic approach for Parkinson's disease. This study aimed to investigate the use of elongated porous gold nanorods (AuNpRs) as an enhancer of cell fate conversion. We observed that AuNpRs promoted the direct conversion of fibroblasts into dopaminergic neurons in vivo and in vitro. The extent of conversion of fibroblasts into dopaminergic neurons depended on the porosity of AuNpRs, as determined by their aspect ratio. The mechanism underlying these results involves specific AuNpR-induced transcriptional changes that altered the expression of antioxidant-related molecules. The generation of dopaminergic neurons via the direct conversion method will open a new avenue for developing a therapeutic platform for Parkinson's disease treatment. STATEMENT OF SIGNIFICANCE: In this study, we applied modified gold nanoporous materials (AuNpRs) to the direct lineage reprogramming of dopaminergic neurons. The cell reprogramming process is energy-intensive, resulting in an excess of oxidative stress. AuNpRs facilitated the direct conversion of dopaminergic neurons by ameliorating oxidative stress during the reprogramming process. We have found this mechanistic clue from high throughput studies in this research work.
Collapse
Affiliation(s)
- Sungwoo Lee
- Department of Chemistry, Sungkyunkwan University, 2066, Seobu-ro, Jangan-gu, Suwon-si, Gyeongki-do, 16419, Republic of Korea
| | - Hyun Soo Shim
- Laboratory of regenerative medicine for neurodegenerative disease, Stand Up Therapeutics, Hannamdaero 98, Seoul, 04418, Republic of Korea; Department of Molecular biology, Nuturn Science, Sinsadong 559-8, Seoul, 06037, Republic of Korea
| | - Hyeok Ju Park
- Laboratory of regenerative medicine for neurodegenerative disease, Stand Up Therapeutics, Hannamdaero 98, Seoul, 04418, Republic of Korea; Database Laboratory, Department of Computer Science and Engineering, Dongguk University-Seoul, Pildong-ro 1-gil 30, Jung-gu, Seoul, 04620, Republic of Korea
| | - Yujung Chang
- Department of Molecular biology, Nuturn Science, Sinsadong 559-8, Seoul, 06037, Republic of Korea
| | - Young-Eun Han
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Soo-Jin Oh
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea; Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology (KIST), Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Wonwoong Lee
- College of Pharmacy, Woosuk University, 443, Samnye-ro, Samnye-eup, Wanju_Gun, Jeollabuk-do, 55338, Republic of Korea
| | - Hyeonjoo Im
- Laboratory of regenerative medicine for neurodegenerative disease, Stand Up Therapeutics, Hannamdaero 98, Seoul, 04418, Republic of Korea; Department of Anatomy, College of Medicine, Korea University, 145, Anam-ro, Seongbuk-gu, Seoul, 20841, Republic of Korea Seoul
| | - YunHee Seol
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Hoon Ryu
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Hoon Kang
- Laboratory of regenerative medicine for neurodegenerative disease, Stand Up Therapeutics, Hannamdaero 98, Seoul, 04418, Republic of Korea; Department of Molecular biology, Nuturn Science, Sinsadong 559-8, Seoul, 06037, Republic of Korea
| | - Yong Kyu Lee
- Database Laboratory, Department of Computer Science and Engineering, Dongguk University-Seoul, Pildong-ro 1-gil 30, Jung-gu, Seoul, 04620, Republic of Korea
| | - Sungho Park
- Department of Chemistry, Sungkyunkwan University, 2066, Seobu-ro, Jangan-gu, Suwon-si, Gyeongki-do, 16419, Republic of Korea.
| | - Junsang Yoo
- Laboratory of regenerative medicine for neurodegenerative disease, Stand Up Therapeutics, Hannamdaero 98, Seoul, 04418, Republic of Korea; Department of Molecular biology, Nuturn Science, Sinsadong 559-8, Seoul, 06037, Republic of Korea.
| |
Collapse
|
38
|
Capano LS, Sato C, Ficulle E, Yu A, Horie K, Kwon JS, Burbach KF, Barthélemy NR, Fox SG, Karch CM, Bateman RJ, Houlden H, Morimoto RI, Holtzman DM, Duff KE, Yoo AS. Recapitulation of endogenous 4R tau expression and formation of insoluble tau in directly reprogrammed human neurons. Cell Stem Cell 2022; 29:918-932.e8. [PMID: 35659876 PMCID: PMC9176216 DOI: 10.1016/j.stem.2022.04.018] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 03/21/2022] [Accepted: 04/27/2022] [Indexed: 01/07/2023]
Abstract
Tau is a microtubule-binding protein expressed in neurons, and the equal ratios between 4-repeat (4R) and 3-repeat (3R) isoforms are maintained in normal adult brain function. Dysregulation of 3R:4R ratio causes tauopathy, and human neurons that recapitulate tau isoforms in health and disease will provide a platform for elucidating pathogenic processes involving tau pathology. We carried out extensive characterizations of tau isoforms expressed in human neurons derived by microRNA-induced neuronal reprogramming of adult fibroblasts. Transcript and protein analyses showed that miR neurons expressed all six isoforms with the 3R:4R isoform ratio equivalent to that detected in human adult brains. Also, miR neurons derived from familial tauopathy patients with a 3R:4R ratio altering mutation showed increased 4R tau and the formation of insoluble tau with seeding activity. Our results collectively demonstrate the utility of miRNA-induced neuronal reprogramming to recapitulate endogenous tau regulation comparable with the adult brain in health and disease.
Collapse
Affiliation(s)
- Lucia S Capano
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Center for Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Program in Molecular and Cell Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Chihiro Sato
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Elena Ficulle
- UK Dementia Research Institute at University College London, London WC1E 6BT, UK
| | - Anan Yu
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, IL 60208, USA
| | - Kanta Horie
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ji-Sun Kwon
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Program in Computational and Systems Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Kyle F Burbach
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Program in Molecular Genetics and Genomics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nicolas R Barthélemy
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Susan G Fox
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, IL 60208, USA
| | - Celeste M Karch
- Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Hope Center for Neurological Disorders, Knight ADRC, St. Louis, MO 63110, USA; Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA; Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Hope Center for Neurological Disorders, Knight ADRC, St. Louis, MO 63110, USA
| | - Henry Houlden
- UK Dementia Research Institute at University College London, London WC1E 6BT, UK
| | - Richard I Morimoto
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, IL 60208, USA
| | - David M Holtzman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA; Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Hope Center for Neurological Disorders, Knight ADRC, St. Louis, MO 63110, USA
| | - Karen E Duff
- UK Dementia Research Institute at University College London, London WC1E 6BT, UK.
| | - Andrew S Yoo
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Center for Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Hope Center for Neurological Disorders, Knight ADRC, St. Louis, MO 63110, USA.
| |
Collapse
|
39
|
Bauer US, Fiskum V, Nair RR, van de Wijdeven R, Kentros C, Sandvig I, Sandvig A. Validation of Functional Connectivity of Engineered Neuromuscular Junction With Recombinant Monosynaptic Pseudotyped ΔG-Rabies Virus Tracing. Front Integr Neurosci 2022; 16:855071. [PMID: 35669734 PMCID: PMC9163662 DOI: 10.3389/fnint.2022.855071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 05/02/2022] [Indexed: 11/15/2022] Open
Abstract
Current preclinical models of neurodegenerative disease, such as amyotrophic lateral sclerosis (ALS), can significantly benefit from in vitro neuroengineering approaches that enable the selective study and manipulation of neurons, networks, and functional units of interest. Custom-designed compartmentalized microfluidic culture systems enable the co-culture of different relevant cell types in interconnected but fluidically isolated microenvironments. Such systems can thus be applied for ALS disease modeling, as they enable the recapitulation and study of neuromuscular junctions (NMJ) through co-culturing of motor neurons and muscle cells in separate, but interconnected compartments. These in vitro systems are particularly relevant for investigations of mechanistic aspects of the ALS pathological cascade in engineered NMJ, as progressive loss of NMJ functionality may constitute one of the hallmarks of disease related pathology at early onset, in line with the dying back hypothesis. In such models, ability to test whether motor neuron degeneration in ALS starts at the nerve terminal or at the NMJ and retrogradely progresses to the motor neuron cell body largely relies on robust methods for verification of engineered NMJ functionality. In this study, we demonstrate the functionality of engineered NMJs within a microfluidic chip with a differentially perturbable microenvironment using a designer pseudotyped ΔG-rabies virus for retrograde monosynaptic tracing.
Collapse
Affiliation(s)
- Ulrich Stefan Bauer
- Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Vegard Fiskum
- Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Rajeevkumar Raveendran Nair
- Centre for Neural Computation, Kavli Institute for Systems Neuroscience, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Rosanne van de Wijdeven
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Clifford Kentros
- Centre for Neural Computation, Kavli Institute for Systems Neuroscience, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Institute of Neuroscience, University of Oregon, Eugene, OR, United States
| | - Ioanna Sandvig
- Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Axel Sandvig
- Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Clinical Neuroscience, Umeå University Hospital, Umeå, Sweden
- Department of Community Medicine and Rehabilitation, Umeå University, Umeå, Sweden
- *Correspondence: Axel Sandvig,
| |
Collapse
|
40
|
Pascale E, Caiazza C, Paladino M, Parisi S, Passaro F, Caiazzo M. MicroRNA Roles in Cell Reprogramming Mechanisms. Cells 2022; 11:940. [PMID: 35326391 PMCID: PMC8946776 DOI: 10.3390/cells11060940] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/28/2022] [Accepted: 03/08/2022] [Indexed: 02/01/2023] Open
Abstract
Cell reprogramming is a groundbreaking technology that, in few decades, generated a new paradigm in biomedical science. To date we can use cell reprogramming to potentially generate every cell type by converting somatic cells and suitably modulating the expression of key transcription factors. This approach can be used to convert skin fibroblasts into pluripotent stem cells as well as into a variety of differentiated and medically relevant cell types, including cardiomyocytes and neural cells. The molecular mechanisms underlying such striking cell phenotypes are still largely unknown, but in the last decade it has been proven that cell reprogramming approaches are significantly influenced by non-coding RNAs. Specifically, this review will focus on the role of microRNAs in the reprogramming processes that lead to the generation of pluripotent stem cells, neurons, and cardiomyocytes. As highlighted here, non-coding RNA-forced expression can be sufficient to support some cell reprogramming processes, and, therefore, we will also discuss how these molecular determinants could be used in the future for biomedical purposes.
Collapse
Affiliation(s)
- Emilia Pascale
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy; (E.P.); (C.C.); (M.P.); (S.P.)
| | - Carmen Caiazza
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy; (E.P.); (C.C.); (M.P.); (S.P.)
| | - Martina Paladino
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy; (E.P.); (C.C.); (M.P.); (S.P.)
| | - Silvia Parisi
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy; (E.P.); (C.C.); (M.P.); (S.P.)
| | - Fabiana Passaro
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy; (E.P.); (C.C.); (M.P.); (S.P.)
| | - Massimiliano Caiazzo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy; (E.P.); (C.C.); (M.P.); (S.P.)
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| |
Collapse
|
41
|
Li W, Li J, Yang Y. Recognition of the Possible miRNA-mRNA Controlling Network in Stroke by Bioinformatics Examination. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2021; 2021:6745282. [PMID: 34938355 PMCID: PMC8687781 DOI: 10.1155/2021/6745282] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/16/2021] [Accepted: 11/05/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Based on the latest research of WHO, it has been revealed that more than 15 million people suffer from stroke every year worldwide. Of these 15 million people, 6 million succumb to death, and 5 million get permanently disabled. This is the prime reason for the substantial economic burden on all parts of the world. METHODS These data have been obtained from the GEO database, and the GEO2R tool was used to find out the differentially expressed miRNAs (DEMs) between the stroke and normal patients' blood. FunRich and miRNet were considered to find potential upstream transcription factors and downstream target genes of candidate EMRs. Next, we use GO annotation and KEGG pathway enrichment. Target genes were analyzed with the help of the R software. Then, the STRING database and Cytoscape software were used to conduct PPI and DEM-hub gene networks. Finally, GSE58294 was used to estimate the hub gene expressions. RESULTS Six DEMs in total were selected out from GSE95204 and GSE117064 datasets. 663 DEMs' target genes were predicted, and NRF1, EGR1, MYC, YY1, E2F1, SP4, and SP1 were predicted as an upstream transcription factor for DEMs' target genes. Target genes of DEMs were primarily augmented in the PI3K-Akt signaling pathway and p53 signaling pathway. The network construction of DEM hygiene is potentially modulated by hsa-miR-3591-5p, hsa-miR-548as-3p, hsa-miR-206, and hsa-miR-4503 hub genes which were found among the top 10 of the hub genes. Among the top 10 hub genes, justification of CTNNB1, PTEN, ESR1, CCND1, KRAS, AKT1, CCND2, CDKN1B, and MYCN was constant with that in the GSE58294 dataset. CONCLUSION In summary, our research first constructs the miRNA-mRNA network in stroke, which probably renders an awakening purview into the pathogenesis and cure of stroke.
Collapse
Affiliation(s)
- Wei Li
- Emergency Department of Benxi Central Hospital, Benxi City, Liaoning Province 117000, China
| | - Jian Li
- Department of Thyroid Head and Neck Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang City, Liaoning Province 110801, China
| | - Yong Yang
- Emergency Department of Benxi Central Hospital, Benxi City, Liaoning Province 117000, China
| |
Collapse
|
42
|
Han F, Liu Y, Huang J, Zhang X, Wei C. Current Approaches and Molecular Mechanisms for Directly Reprogramming Fibroblasts Into Neurons and Dopamine Neurons. Front Aging Neurosci 2021; 13:738529. [PMID: 34658841 PMCID: PMC8515543 DOI: 10.3389/fnagi.2021.738529] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 08/27/2021] [Indexed: 12/30/2022] Open
Abstract
Parkinson's disease is mainly caused by specific degeneration of dopaminergic neurons (DA neurons) in the substantia nigra of the middle brain. Over the past two decades, transplantation of neural stem cells (NSCs) from fetal brain-derived neural stem cells (fNSCs), human embryonic stem cells (hESCs), and induced pluripotent stem cells (iPSCs) has been shown to improve the symptoms of motor dysfunction in Parkinson's disease (PD) animal models and PD patients significantly. However, there are ethical concerns with fNSCs and hESCs and there is an issue of rejection by the immune system, and the iPSCs may involve tumorigenicity caused by the integration of the transgenes. Recent studies have shown that somatic fibroblasts can be directly reprogrammed to NSCs, neurons, and specific dopamine neurons. Directly induced neurons (iN) or induced DA neurons (iDANs) from somatic fibroblasts have several advantages over iPSC cells. The neurons produced by direct transdifferentiation do not pass through a pluripotent state. Therefore, direct reprogramming can generate patient-specific cells, and it can overcome the safety problems of rejection by the immune system and teratoma formation related to hESCs and iPSCs. However, there are some critical issues such as the low efficiency of direct reprogramming, biological functions, and risks from the directly converted neurons, which hinder their clinical applications. Here, the recent progress in methods, mechanisms, and future challenges of directly reprogramming somatic fibroblasts into neurons or dopamine neurons were summarized to speed up the clinical translation of these directly converted neural cells to treat PD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Fabin Han
- Innovation Institute for Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.,Shenzhen Research Institute of Shandong University, Jinan, China.,The Institute for Tissue Engineering and Regenerative Medicine, Liaocheng University/Liaocheng People's Hospital, Liaocheng, China
| | - Yanming Liu
- Shenzhen Research Institute of Shandong University, Jinan, China.,The Institute for Tissue Engineering and Regenerative Medicine, Liaocheng University/Liaocheng People's Hospital, Liaocheng, China
| | - Jin Huang
- Laboratory of Basic Medical Research, Medical Centre of PLA Strategic Support Force, Beijing, China
| | - Xiaoping Zhang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chuanfei Wei
- The Institute for Tissue Engineering and Regenerative Medicine, Liaocheng University/Liaocheng People's Hospital, Liaocheng, China
| |
Collapse
|
43
|
Samoilova EM, Belopasov VV, Baklaushev VP. Transcription Factors of Direct Neuronal Reprogramming in Ontogenesis and Ex Vivo. Mol Biol 2021; 55:645-669. [DOI: 10.1134/s0026893321040087] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 03/07/2025]
|
44
|
Cho J, Kim S, Lee H, Rah W, Cho HC, Kim NK, Bae S, Shin DH, Lee MG, Park IH, Tanaka Y, Shin E, Yi H, Han JW, Hwang PTJ, Jun HW, Park HJ, Cho K, Lee SW, Jung JK, Levit RD, Sussman MA, Harvey RP, Yoon YS. Regeneration of infarcted mouse hearts by cardiovascular tissue formed via the direct reprogramming of mouse fibroblasts. Nat Biomed Eng 2021; 5:880-896. [PMID: 34426676 PMCID: PMC8809198 DOI: 10.1038/s41551-021-00783-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 07/13/2021] [Indexed: 02/07/2023]
Abstract
Fibroblasts can be directly reprogrammed into cardiomyocytes, endothelial cells or smooth muscle cells. Here we report the reprogramming of mouse tail-tip fibroblasts simultaneously into cells resembling these three cell types using the microRNA mimic miR-208b-3p, ascorbic acid and bone morphogenetic protein 4, as well as the formation of tissue-like structures formed by the directly reprogrammed cells. Implantation of the formed cardiovascular tissue into the infarcted hearts of mice led to the migration of reprogrammed cells to the injured tissue, reducing regional cardiac strain and improving cardiac function. The migrated endothelial cells and smooth muscle cells contributed to vessel formation, and the migrated cardiomyocytes, which initially displayed immature characteristics, became mature over time and formed gap junctions with host cardiomyocytes. Direct reprogramming of somatic cells to make cardiac tissue may aid the development of applications in cell therapy, disease modelling and drug discovery for cardiovascular diseases.
Collapse
Affiliation(s)
- Jaeyeaon Cho
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sangsung Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyein Lee
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Woongchan Rah
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hee Cheol Cho
- Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Nam Kyun Kim
- Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Seongho Bae
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Dong Hoon Shin
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Min Goo Lee
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - In-Hyun Park
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT, USA
| | - Yoshiaki Tanaka
- Department of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Eric Shin
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Hong Yi
- Robert P. Apkarian Integrated Electron Microscopy Core, Emory University, Atlanta, GA, USA
| | - Ji Woong Han
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Patrick Tae Joon Hwang
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ho-Wook Jun
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hun-Jun Park
- Division of Cardiology, Department of Internal Medicine, Seoul St Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kyuwon Cho
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Sang Wook Lee
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Jae Kyung Jung
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Rebecca D Levit
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Mark A Sussman
- San Diego State University Heart Institute, San Diego State University, San Diego, CA, USA
- Department of Biology, San Diego State University, San Diego, CA, USA
| | - Richard P Harvey
- Victor Chang Cardiac Research Institute, St Vincent's Hospital, Darlinghurst, New South Wales, Australia
| | - Young-Sup Yoon
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA.
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
45
|
Trudler D, Ghatak S, Lipton SA. Emerging hiPSC Models for Drug Discovery in Neurodegenerative Diseases. Int J Mol Sci 2021; 22:8196. [PMID: 34360966 PMCID: PMC8347370 DOI: 10.3390/ijms22158196] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 12/12/2022] Open
Abstract
Neurodegenerative diseases affect millions of people worldwide and are characterized by the chronic and progressive deterioration of neural function. Neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and Huntington's disease (HD), represent a huge social and economic burden due to increasing prevalence in our aging society, severity of symptoms, and lack of effective disease-modifying therapies. This lack of effective treatments is partly due to a lack of reliable models. Modeling neurodegenerative diseases is difficult because of poor access to human samples (restricted in general to postmortem tissue) and limited knowledge of disease mechanisms in a human context. Animal models play an instrumental role in understanding these diseases but fail to comprehensively represent the full extent of disease due to critical differences between humans and other mammals. The advent of human-induced pluripotent stem cell (hiPSC) technology presents an advantageous system that complements animal models of neurodegenerative diseases. Coupled with advances in gene-editing technologies, hiPSC-derived neural cells from patients and healthy donors now allow disease modeling using human samples that can be used for drug discovery.
Collapse
Affiliation(s)
- Dorit Trudler
- Neurodegeneration New Medicines Center and Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA; (D.T.); (S.G.)
| | - Swagata Ghatak
- Neurodegeneration New Medicines Center and Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA; (D.T.); (S.G.)
| | - Stuart A. Lipton
- Neurodegeneration New Medicines Center and Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA; (D.T.); (S.G.)
- Department of Neurosciences, University of California San Diego School of Medicine, La Jolla, CA 92093, USA
| |
Collapse
|
46
|
Kempf J, Knelles K, Hersbach BA, Petrik D, Riedemann T, Bednarova V, Janjic A, Simon-Ebert T, Enard W, Smialowski P, Götz M, Masserdotti G. Heterogeneity of neurons reprogrammed from spinal cord astrocytes by the proneural factors Ascl1 and Neurogenin2. Cell Rep 2021; 36:109409. [PMID: 34289357 PMCID: PMC8316252 DOI: 10.1016/j.celrep.2021.109409] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/14/2021] [Accepted: 06/24/2021] [Indexed: 01/21/2023] Open
Abstract
Astrocytes are a viable source for generating new neurons via direct conversion. However, little is known about the neurogenic cascades triggered in astrocytes from different regions of the CNS. Here, we examine the transcriptome induced by the proneural factors Ascl1 and Neurog2 in spinal cord-derived astrocytes in vitro. Each factor initially elicits different neurogenic programs that later converge to a V2 interneuron-like state. Intriguingly, patch sequencing (patch-seq) shows no overall correlation between functional properties and the transcriptome of the heterogenous induced neurons, except for K-channels. For example, some neurons with fully mature electrophysiological properties still express astrocyte genes, thus calling for careful molecular and functional analysis. Comparing the transcriptomes of spinal cord- and cerebral-cortex-derived astrocytes reveals profound differences, including developmental patterning cues maintained in vitro. These relate to the distinct neuronal identity elicited by Ascl1 and Neurog2 reflecting their developmental functions in subtype specification of the respective CNS region.
Collapse
Affiliation(s)
- J Kempf
- Biomedical Center Munich, Physiological Genomics, LMU Munich, Planegg-Martinsried 82152, Germany
| | - K Knelles
- Biomedical Center Munich, Physiological Genomics, LMU Munich, Planegg-Martinsried 82152, Germany
| | - B A Hersbach
- Biomedical Center Munich, Physiological Genomics, LMU Munich, Planegg-Martinsried 82152, Germany; Institute for Stem Cell Research, Helmholtz Center Munich, Neuherberg 85764, Germany; Graduate School of Systemic Neurosciences, LMU Munich, Planegg-Martinsried 82152, Germany
| | - D Petrik
- Biomedical Center Munich, Physiological Genomics, LMU Munich, Planegg-Martinsried 82152, Germany; Institute for Stem Cell Research, Helmholtz Center Munich, Neuherberg 85764, Germany; School of Biosciences, The Sir Martin Evans Building, Cardiff University, CF10 3AX Cardiff, UK
| | - T Riedemann
- Biomedical Center Munich, Physiological Genomics, LMU Munich, Planegg-Martinsried 82152, Germany
| | - V Bednarova
- Biomedical Center Munich, Physiological Genomics, LMU Munich, Planegg-Martinsried 82152, Germany
| | - A Janjic
- Anthropology and Human Genomics, Faculty of Biology, LMU Munich, Planegg-Martinsried 82152, Germany
| | - T Simon-Ebert
- Biomedical Center Munich, Physiological Genomics, LMU Munich, Planegg-Martinsried 82152, Germany
| | - W Enard
- Biomedical Center Munich, Bioinformatic Core Facility, LMU Munich, Planegg-Martinsried 82152, Germany
| | - P Smialowski
- Biomedical Center Munich, Physiological Genomics, LMU Munich, Planegg-Martinsried 82152, Germany; Institute for Stem Cell Research, Helmholtz Center Munich, Neuherberg 85764, Germany; School of Biosciences, The Sir Martin Evans Building, Cardiff University, CF10 3AX Cardiff, UK
| | - M Götz
- Biomedical Center Munich, Physiological Genomics, LMU Munich, Planegg-Martinsried 82152, Germany; Institute for Stem Cell Research, Helmholtz Center Munich, Neuherberg 85764, Germany; Excellence Cluster of Systems Neurology (SYNERGY), Munich, Germany.
| | - G Masserdotti
- Biomedical Center Munich, Physiological Genomics, LMU Munich, Planegg-Martinsried 82152, Germany; Institute for Stem Cell Research, Helmholtz Center Munich, Neuherberg 85764, Germany.
| |
Collapse
|
47
|
Zeng Y, Li N, Zheng Z, Chen R, Liu W, Cheng J, Zhu J, Zeng M, Peng M, Hong C. Screening of key biomarkers and immune infiltration in Pulmonary Arterial Hypertension via integrated bioinformatics analysis. Bioengineered 2021; 12:2576-2591. [PMID: 34233597 PMCID: PMC8806790 DOI: 10.1080/21655979.2021.1936816] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
This study aimed to screen key biomarkers and investigate immune infiltration in pulmonary arterial hypertension (PAH) based on integrated bioinformatics analysis. The Gene Expression Omnibus (GEO) database was used to download three mRNA expression profiles comprising 91 PAH lung specimens and 49 normal lung specimens. Three mRNA expression datasets were combined, and differentially expressed genes (DEGs) were obtained. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses and the protein-protein interaction (PPI) network of DEGs were performed using the STRING and DAVID databases, respectively. The diagnostic value of hub gene expression in PAH was also analyzed. Finally, the infiltration of immune cells in PAH was analyzed using the CIBERSORT algorithm. Total 182 DEGs (117 upregulated and 65 downregulated) were identified, and 15 hub genes were screened. These 15 hub genes were significantly associated with immune system functions such as myeloid leukocyte migration, neutrophil migration, cell chemotaxis, Toll-like receptor signaling pathway, and NF-κB signaling pathway. A 7-gene-based model was constructed and had a better diagnostic value in identifying PAH tissues compared with normal controls. The immune infiltration profiles of the PAH and normal control samples were significantly different. High proportions of resting NK cells, activated mast cells, monocytes, and neutrophils were found in PAH samples, while high proportions of resting T cells CD4 memory and Macrophages M1 cell were found in normal control samples. Functional enrichment of DEGs and immune infiltration analysis between PAH and normal control samples might help to understand the pathogenesis of PAH.
Collapse
Affiliation(s)
- Yu Zeng
- Department of Respiration, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Nanhong Li
- Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Zhenzhen Zheng
- Department of Respiration, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Riken Chen
- China State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wang Liu
- Department of Respiration, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Junfen Cheng
- Department of Respiration, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Jinru Zhu
- Department of Respiration, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Mingqing Zeng
- First Clinical School of Medicine, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Min Peng
- Department of Respiration, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Cheng Hong
- China State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
48
|
Vasan L, Park E, David LA, Fleming T, Schuurmans C. Direct Neuronal Reprogramming: Bridging the Gap Between Basic Science and Clinical Application. Front Cell Dev Biol 2021; 9:681087. [PMID: 34291049 PMCID: PMC8287587 DOI: 10.3389/fcell.2021.681087] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 06/02/2021] [Indexed: 12/15/2022] Open
Abstract
Direct neuronal reprogramming is an innovative new technology that involves the conversion of somatic cells to induced neurons (iNs) without passing through a pluripotent state. The capacity to make new neurons in the brain, which previously was not achievable, has created great excitement in the field as it has opened the door for the potential treatment of incurable neurodegenerative diseases and brain injuries such as stroke. These neurological disorders are associated with frank neuronal loss, and as new neurons are not made in most of the adult brain, treatment options are limited. Developmental biologists have paved the way for the field of direct neuronal reprogramming by identifying both intrinsic cues, primarily transcription factors (TFs) and miRNAs, and extrinsic cues, including growth factors and other signaling molecules, that induce neurogenesis and specify neuronal subtype identities in the embryonic brain. The striking observation that postmitotic, terminally differentiated somatic cells can be converted to iNs by mis-expression of TFs or miRNAs involved in neural lineage development, and/or by exposure to growth factors or small molecule cocktails that recapitulate the signaling environment of the developing brain, has opened the door to the rapid expansion of new neuronal reprogramming methodologies. Furthermore, the more recent applications of neuronal lineage conversion strategies that target resident glial cells in situ has expanded the clinical potential of direct neuronal reprogramming techniques. Herein, we present an overview of the history, accomplishments, and therapeutic potential of direct neuronal reprogramming as revealed over the last two decades.
Collapse
Affiliation(s)
- Lakshmy Vasan
- Sunnybrook Research Institute, Biological Sciences Platform, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Eunjee Park
- Sunnybrook Research Institute, Biological Sciences Platform, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Luke Ajay David
- Sunnybrook Research Institute, Biological Sciences Platform, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Taylor Fleming
- Sunnybrook Research Institute, Biological Sciences Platform, Toronto, ON, Canada
| | - Carol Schuurmans
- Sunnybrook Research Institute, Biological Sciences Platform, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
49
|
MiR-124 synergism with ELAVL3 enhances target gene expression to promote neuronal maturity. Proc Natl Acad Sci U S A 2021; 118:2015454118. [PMID: 34031238 DOI: 10.1073/pnas.2015454118] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Neuron-enriched microRNAs (miRNAs), miR-9/9* and miR-124 (miR-9/9*-124), direct cell fate switching of human fibroblasts to neurons when ectopically expressed by repressing antineurogenic genes. How these miRNAs function after the repression of fibroblast genes for neuronal fate remains unclear. Here, we identified targets of miR-9/9*-124 as reprogramming cells activate the neuronal program and reveal the role of miR-124 that directly promotes the expression of its target genes associated with neuronal development and function. The mode of miR-124 as a positive regulator is determined by the binding of both AGO and a neuron-enriched RNA-binding protein, ELAVL3, to target transcripts. Although existing literature indicates that miRNA-ELAVL family protein interaction can result in either target gene up-regulation or down-regulation in a context-dependent manner, we specifically identified neuronal ELAVL3 as the driver for miR-124 target gene up-regulation in neurons. In primary human neurons, repressing miR-124 and ELAVL3 led to the down-regulation of genes involved in neuronal function and process outgrowth and cellular phenotypes of reduced inward currents and neurite outgrowth. Our results highlight the synergistic role between miR-124 and RNA-binding proteins to promote target gene regulation and neuronal function.
Collapse
|
50
|
Abstract
Direct cell fate conversion of human somatic cells into induced neurons (iNs) is often regarded as a highly concerted one-step process. In this issue of Cell Stem Cell, Cates et al. (2021) dissect the iN conversion trajectory into two largely independent steps and identify key players at each stage.
Collapse
Affiliation(s)
- Joseph R Herdy
- Neural Aging Laboratory, Institute of Molecular Biology, CMBI, Leopold-Franzens-University Innsbruck, Austria; Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Lukas Karbacher
- Neural Aging Laboratory, Institute of Molecular Biology, CMBI, Leopold-Franzens-University Innsbruck, Austria
| | - Jerome Mertens
- Neural Aging Laboratory, Institute of Molecular Biology, CMBI, Leopold-Franzens-University Innsbruck, Austria; Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA.
| |
Collapse
|