Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Biol Evol ; 39(10)2022 10 07.
Article in English | MEDLINE | ID: mdl-36205081

ABSTRACT

Although new genes can arrive from modes other than duplication, few examples are well characterized. Given high expression in some human brain subregions and a putative link to psychological disorders [e.g., schizophrenia (SCZ)], suggestive of brain functionality, here we characterize piggyBac transposable element-derived 1 (PGBD1). PGBD1 is nonmonotreme mammal-specific and under purifying selection, consistent with functionality. The gene body of human PGBD1 retains much of the original DNA transposon but has additionally captured SCAN and KRAB domains. Despite gene body retention, PGBD1 has lost transposition abilities, thus transposase functionality is absent. PGBD1 no longer recognizes piggyBac transposon-like inverted repeats, nonetheless PGBD1 has DNA binding activity. Genome scale analysis identifies enrichment of binding sites in and around genes involved in neuronal development, with association with both histone activating and repressing marks. We focus on one of the repressed genes, the long noncoding RNA NEAT1, also dysregulated in SCZ, the core structural RNA of paraspeckles. DNA binding assays confirm specific binding of PGBD1 both in the NEAT1 promoter and in the gene body. Depletion of PGBD1 in neuronal progenitor cells (NPCs) results in increased NEAT1/paraspeckles and differentiation. We conclude that PGBD1 has evolved core regulatory functionality for the maintenance of NPCs. As paraspeckles are a mammal-specific structure, the results presented here show a rare example of the evolution of a novel gene coupled to the evolution of a contemporaneous new structure.


Subject(s)
DNA Transposable Elements , RNA, Long Noncoding , Animals , Cell Nucleus/genetics , Histones/metabolism , Humans , Mammals/genetics , Mammals/metabolism , Nerve Tissue Proteins , Paraspeckles , RNA, Long Noncoding/metabolism , Transposases/genetics , Transposases/metabolism
2.
J Vis Exp ; (172)2021 06 21.
Article in English | MEDLINE | ID: mdl-34223837

ABSTRACT

Mitochondrial diseases represent the largest class of inborn errors of metabolism and are currently incurable. These diseases cause neurodevelopmental defects whose underlying mechanisms remain to be elucidated. A major roadblock is the lack of effective models recapitulating the early-onset neuronal impairment seen in the patients. Advances in the technology of induced pluripotent stem cells (iPSCs) enable the generation of three-dimensional (3D) brain organoids that can be used to investigate the impact of diseases on the development and organization of the nervous system. Researchers, including these authors, have recently introduced human brain organoids to model mitochondrial disorders. This paper reports a detailed protocol for the robust generation of human iPSC-derived brain organoids and their use in mitochondrial bioenergetic profiling and imaging analyses. These experiments will allow the use of brain organoids to investigate metabolic and developmental dysfunctions and may provide crucial information to dissect the neuronal pathology of mitochondrial diseases.


Subject(s)
Induced Pluripotent Stem Cells , Mitochondrial Diseases , Brain , Cell Differentiation , Humans , Organoids
3.
Methods Mol Biol ; 2277: 391-403, 2021.
Article in English | MEDLINE | ID: mdl-34080164

ABSTRACT

Cellular metabolism contributes to cell fate decisions. Bioenergetic profiling can therefore provide considerable insights into cellular identity and specification. Given the current importance of human pluripotent stem cells (hPSCs) for biomedical applications, assessing the bioenergetic properties of hPSCs and derivatives can unveil relevant mechanisms in the context of development biology and molecular disease modeling. Here, we describe a method to facilitate bioenergetic profiling of hPSCs in a reproducible and scalable manner. After simultaneous assessment of mitochondrial respiration and glycolytic capacity using Seahorse XFe96 Analyzer, we measure lactate concentration in the cellular media. Finally, we normalize the values based on DNA amount. We describe the procedures with specific requirements related to hPSCs . However, the same protocol can be easily adapted to other cell types, including differentiated progenies from hPSCs .


Subject(s)
Mitochondria/metabolism , Molecular Biology/methods , Pluripotent Stem Cells/metabolism , Antimycin A/pharmacology , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/pharmacology , Cell Culture Techniques/methods , DNA/analysis , Energy Metabolism/drug effects , Humans , Lactic Acid/analysis , Mitochondria/drug effects , Oligomycins/pharmacology , Oxygen Consumption/drug effects , Pluripotent Stem Cells/drug effects , Rotenone/pharmacology
4.
Nat Commun ; 12(1): 1929, 2021 03 26.
Article in English | MEDLINE | ID: mdl-33771987

ABSTRACT

Leigh syndrome (LS) is a severe manifestation of mitochondrial disease in children and is currently incurable. The lack of effective models hampers our understanding of the mechanisms underlying the neuronal pathology of LS. Using patient-derived induced pluripotent stem cells and CRISPR/Cas9 engineering, we developed a human model of LS caused by mutations in the complex IV assembly gene SURF1. Single-cell RNA-sequencing and multi-omics analysis revealed compromised neuronal morphogenesis in mutant neural cultures and brain organoids. The defects emerged at the level of neural progenitor cells (NPCs), which retained a glycolytic proliferative state that failed to instruct neuronal morphogenesis. LS NPCs carrying mutations in the complex I gene NDUFS4 recapitulated morphogenesis defects. SURF1 gene augmentation and PGC1A induction via bezafibrate treatment supported the metabolic programming of LS NPCs, leading to restored neuronal morphogenesis. Our findings provide mechanistic insights and suggest potential interventional strategies for a rare mitochondrial disease.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Leigh Disease/genetics , Membrane Proteins/genetics , Mitochondrial Proteins/genetics , Mutation , Neurons/metabolism , Organoids/metabolism , Cells, Cultured , Child, Preschool , Humans , Induced Pluripotent Stem Cells/cytology , Leigh Disease/metabolism , Male , Metabolomics/methods , Mitochondria/genetics , Mitochondria/metabolism , Morphogenesis/genetics , Neurons/cytology , Proteomics/methods , Single-Cell Analysis/methods , Exome Sequencing
5.
Stem Cells ; 35(7): 1655-1662, 2017 07.
Article in English | MEDLINE | ID: mdl-28544378

ABSTRACT

High attrition rates and loss of capital plague the drug discovery process. This is particularly evident for mitochondrial disease that typically involves neurological manifestations and is caused by nuclear or mitochondrial DNA defects. This group of heterogeneous disorders is difficult to target because of the variability of the symptoms among individual patients and the lack of viable modeling systems. The use of induced pluripotent stem cells (iPSCs) might significantly improve the search for effective therapies for mitochondrial disease. iPSCs can be used to generate patient-specific neural cell models in which innovative compounds can be identified or validated. Here we discuss the promises and challenges of iPSC-based drug discovery for mitochondrial disease with a specific focus on neurological conditions. We anticipate that a proper use of the potent iPSC technology will provide critical support for the development of innovative therapies against these untreatable and detrimental disorders. Stem Cells 2017;35:1655-1662.


Subject(s)
Drug Discovery/methods , Induced Pluripotent Stem Cells/drug effects , Mitochondria/drug effects , Mitochondrial Diseases/drug therapy , Neurons/drug effects , Neuroprotective Agents/pharmacology , Cell Differentiation , DNA, Mitochondrial/genetics , Gene Expression , High-Throughput Screening Assays , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Mitochondrial Proteins/agonists , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Models, Biological , Mutation , Neurons/metabolism , Neurons/pathology , Organoids/drug effects , Organoids/metabolism , Organoids/pathology , Precision Medicine
6.
Cell Stem Cell ; 20(5): 659-674.e9, 2017 05 04.
Article in English | MEDLINE | ID: mdl-28132834

ABSTRACT

Mitochondrial DNA (mtDNA) mutations frequently cause neurological diseases. Modeling of these defects has been difficult because of the challenges associated with engineering mtDNA. We show here that neural progenitor cells (NPCs) derived from human induced pluripotent stem cells (iPSCs) retain the parental mtDNA profile and exhibit a metabolic switch toward oxidative phosphorylation. NPCs derived in this way from patients carrying a deleterious homoplasmic mutation in the mitochondrial gene MT-ATP6 (m.9185T>C) showed defective ATP production and abnormally high mitochondrial membrane potential (MMP), plus altered calcium homeostasis, which represents a potential cause of neural impairment. High-content screening of FDA-approved drugs using the MMP phenotype highlighted avanafil, which we found was able to partially rescue the calcium defect in patient NPCs and differentiated neurons. Overall, our results show that iPSC-derived NPCs provide an effective model for drug screening to target mtDNA disorders that affect the nervous system.


Subject(s)
DNA, Mitochondrial/genetics , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Mitochondria/genetics , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Calcium/metabolism , Cell Line , Drug Discovery/methods , Humans , Mutation
7.
Semin Cell Dev Biol ; 52: 102-9, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26877213

ABSTRACT

The metabolic switch associated with the reprogramming of somatic cells to pluripotency has received increasing attention in recent years. However, the impact of mitochondrial and metabolic modulation on stem cell differentiation into neuronal/glial cells and related brain disease modeling still remains to be fully addressed. Here, we seek to focus on this aspect by first addressing brain energy metabolism and its inter-cellular metabolic compartmentalization. We then review the findings related to the mitochondrial and metabolic reconfiguration occurring upon neuronal/glial specification from pluripotent stem cells (PSCs). Finally, we provide an update of the PSC-based models of mitochondria-related brain disorders and discuss the challenges and opportunities that may exist on the road to develop a new era of brain disease modeling and therapy.


Subject(s)
Brain Diseases/metabolism , Induced Pluripotent Stem Cells/metabolism , Neuroglia/metabolism , Neurons/metabolism , Animals , Brain Diseases/pathology , Cell Differentiation/physiology , Energy Metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Neuroglia/cytology , Neurons/cytology
8.
Brain ; 138(Pt 7): 1843-62, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25981963

ABSTRACT

Regeneration of injured central nervous system axons is highly restricted, causing neurological impairment. To date, although the lack of intrinsic regenerative potential is well described, a key regulatory molecular mechanism for the enhancement of both axonal regrowth and functional recovery after central nervous system injury remains elusive. While ubiquitin ligases coordinate neuronal morphogenesis and connectivity during development as well as after axonal injury, their role specifically in axonal regeneration is unknown. Following a bioinformatics network analysis combining ubiquitin ligases with previously defined axonal regenerative proteins, we found a triad composed of the ubiquitin ligases MDM4, MDM2 and the transcription factor p53 (encoded by TP53) as a putative central signalling complex restricting the regeneration program. Indeed, conditional deletion of MDM4 or pharmacological inhibition of MDM2/p53 interaction in the eye and spinal cord promote axonal regeneration and sprouting of the optic nerve after crush and of supraspinal tracts after spinal cord injury. The double conditional deletion of MDM4-p53 as well as MDM2 inhibition in p53-deficient mice blocks this regenerative phenotype, showing its dependence upon p53. Genome-wide gene expression analysis from ex vivo fluorescence-activated cell sorting in MDM4-deficient retinal ganglion cells identifies the downstream target IGF1R, whose activity and expression was found to be required for the regeneration elicited by MDM4 deletion. Importantly, we demonstrate that pharmacological enhancement of the MDM2/p53-IGF1R axis enhances axonal sprouting as well as functional recovery after spinal cord injury. Thus, our results show MDM4-MDM2/p53-IGF1R as an original regulatory mechanism for CNS regeneration and offer novel targets to enhance neurological recovery.media-1vid110.1093/brain/awv125_video_abstractawv125_video_abstract.


Subject(s)
Nerve Regeneration/physiology , Optic Nerve Injuries/metabolism , Recovery of Function/physiology , Signal Transduction/physiology , Spinal Cord Injuries/metabolism , Animals , Axons/metabolism , Axons/pathology , Computational Biology , Disease Models, Animal , Flow Cytometry , Immunoblotting , Immunohistochemistry , Immunoprecipitation , Insulin-Like Growth Factor I/metabolism , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Nerve Crush , Optic Nerve Injuries/pathology , Optic Nerve Injuries/physiopathology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , Transcriptome , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...