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











Publication year range
2.
Biology (Basel) ; 13(5)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38785794

ABSTRACT

The hair follicle is a specialized appendage of the skin that is critical for multiple functions, including thermoregulation, immune surveillance, and sebum production. Mammals are born with a fixed number of hair follicles that develop embryonically. Postnatally, these hair follicles undergo regenerative cycles of regression and growth that recapitulate many of the embryonic signaling pathways. Furthermore, hair cycles have a direct impact on skin regeneration in homeostasis, cutaneous wound healing, and disease conditions such as alopecia. Here, we review the current knowledge of hair follicle formation during embryonic development and the post-natal hair cycle, with an emphasis on the molecular signaling pathways underlying these processes. We then discuss efforts to capitalize on the field's understanding of in vivo mechanisms to bioengineer hair follicles or hair-bearing skin in vitro and how such models may be further improved to develop strategies for hair regeneration.

3.
Br J Dermatol ; 188(2): 259-267, 2023 02 10.
Article in English | MEDLINE | ID: mdl-36763876

ABSTRACT

BACKGROUND: A trio exome sequencing study identified a previously unreported NLRP1 gene variant resulting in a p.Leu813Pro substitution of the LRR (leucine-rich repeats) domain of the NLRP1 protein (NACHT, LRR and PYD domains-containing protein 1). This homozygous mutation was shared by two sisters with different clinical presentation: the younger sister had generalized inflammatory nodules with keratotic plugs, clinically resembling multiple keratoacanthomas, while the older had manifestations of familial keratosis lichenoides chronica. OBJECTIVES: To analyse the consequences of this NLRP1 variant in two siblings with a different clinical spectrum of severity. METHODS: To demonstrate the pathogenicity, p.Leu813Pro was recombinantly expressed, and its effect on inflammasome assembly was assessed. Exome sequencing and RNA-Seq were performed to identify factors with potentially modifying effects on the severity of the skin manifestation between each sibling. RESULTS: The variant p.Leu813Pro triggered activation of the NLRP1 inflammasome leading to ASC (apoptosis-associated speck-like protein containing a CARD) speck formation and interleukin (IL)-1ß release. The more severely affected sister had several additional genomic variants associated with atopy and psoriasis that were not present in her sibling. IL-5 and IL-17 emerged as dominant cytokines driving prominent inflammation in the skin of the severely affected sibling. CONCLUSIONS: To the best of our knowledge, this is the first report of a NLRP1 variant that leads to a different clinical spectrum of severity within the same sibship. IL-5 and IL-17 were the main cytokines expressed in the inflammatory lesions of the severely affected patient and might be regarded as disease modifying factors, and therefore may be considered as therapeutic targets.


Subject(s)
Apoptosis Regulatory Proteins , Inflammasomes , Female , Humans , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Cytokines/metabolism , Gain of Function Mutation , Inflammasomes/metabolism , Interleukin-17/metabolism , Interleukin-5/genetics , Interleukin-5/metabolism , NLR Proteins/genetics , NLR Proteins/metabolism , Phenotype , Siblings
4.
Sci Immunol ; 7(75): eabi4611, 2022 09 16.
Article in English | MEDLINE | ID: mdl-36112693

ABSTRACT

Dipeptidyl peptidase 9 (DPP9) is a direct inhibitor of NLRP1, but how it affects inflammasome regulation in vivo is not yet established. Here, we report three families with immune-associated defects, poor growth, pancytopenia, and skin pigmentation abnormalities that segregate with biallelic DPP9 rare variants. Using patient-derived primary cells and biochemical assays, these variants were shown to behave as hypomorphic or knockout alleles that failed to repress NLRP1. The removal of a single copy of Nlrp1a/b/c, Asc, Gsdmd, or Il-1r, but not Il-18, was sufficient to rescue the lethality of Dpp9 mutant neonates in mice. Similarly, dpp9 deficiency was partially rescued by the inactivation of asc, an obligate downstream adapter of the NLRP1 inflammasome, in zebrafish. These experiments suggest that the deleterious consequences of DPP9 deficiency were mostly driven by the aberrant activation of the canonical NLRP1 inflammasome and IL-1ß signaling. Collectively, our results delineate a Mendelian disorder of DPP9 deficiency driven by increased NLRP1 activity as demonstrated in patient cells and in two animal models of the disease.


Subject(s)
Apoptosis Regulatory Proteins , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases , Inflammasomes , Animals , Mice , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/genetics , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Inflammasomes/metabolism , Interleukin-1/metabolism , NLR Proteins/genetics , Zebrafish
5.
Nat Genet ; 54(8): 1214-1226, 2022 08.
Article in English | MEDLINE | ID: mdl-35864190

ABSTRACT

Cirrhosis is usually a late-onset and life-threatening disease characterized by fibrotic scarring and inflammation that disrupts liver architecture and function. While it is typically the result of alcoholism or hepatitis viral infection in adults, its etiology in infants is much less understood. In this study, we report 14 children from ten unrelated families presenting with a syndromic form of pediatric liver cirrhosis. By genome/exome sequencing, we found recessive variants in FOCAD segregating with the disease. Zebrafish lacking focad phenocopied the human disease, revealing a signature of altered messenger RNA (mRNA) degradation processes in the liver. Using patient's primary cells and CRISPR-Cas9-mediated inactivation in human hepatic cell lines, we found that FOCAD deficiency compromises the SKI mRNA surveillance pathway by reducing the levels of the RNA helicase SKIC2 and its cofactor SKIC3. FOCAD knockout hepatocytes exhibited lowered albumin expression and signs of persistent injury accompanied by CCL2 overproduction. Our results reveal the importance of FOCAD in maintaining liver homeostasis and disclose a possible therapeutic intervention point via inhibition of the CCL2/CCR2 signaling axis.


Subject(s)
Liver Cirrhosis , Tumor Suppressor Proteins , Adult , Animals , Child , Hepatocytes/metabolism , Humans , Liver/metabolism , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Syndrome , Tumor Suppressor Proteins/genetics , Zebrafish/genetics
6.
Mol Cell ; 82(13): 2385-2400.e9, 2022 07 07.
Article in English | MEDLINE | ID: mdl-35594856

ABSTRACT

Inflammation observed in SARS-CoV-2-infected patients suggests that inflammasomes, proinflammatory intracellular complexes, regulate various steps of infection. Lung epithelial cells express inflammasome-forming sensors and constitute the primary entry door of SARS-CoV-2. Here, we describe that the NLRP1 inflammasome detects SARS-CoV-2 infection in human lung epithelial cells. Specifically, human NLRP1 is cleaved at the Q333 site by multiple coronavirus 3CL proteases, which triggers inflammasome assembly and cell death and limits the production of infectious viral particles. Analysis of NLRP1-associated pathways unveils that 3CL proteases also inactivate the pyroptosis executioner Gasdermin D (GSDMD). Subsequently, caspase-3 and GSDME promote alternative cell pyroptosis. Finally, analysis of pyroptosis markers in plasma from COVID-19 patients with characterized severe pneumonia due to autoantibodies against, or inborn errors of, type I interferons (IFNs) highlights GSDME/caspase-3 as potential markers of disease severity. Overall, our findings identify NLRP1 as a sensor of SARS-CoV-2 infection in lung epithelia.


Subject(s)
COVID-19 , Coronavirus 3C Proteases , Epithelial Cells , Inflammasomes , NLR Proteins , SARS-CoV-2 , COVID-19/genetics , COVID-19/metabolism , COVID-19/virology , Caspase 3/metabolism , Coronavirus 3C Proteases/genetics , Coronavirus 3C Proteases/metabolism , Epithelial Cells/metabolism , Humans , Inflammasomes/genetics , Inflammasomes/metabolism , Lung/metabolism , Lung/virology , NLR Proteins/genetics , NLR Proteins/metabolism , Peptide Hydrolases/genetics , Peptide Hydrolases/metabolism , Phosphate-Binding Proteins/genetics , Phosphate-Binding Proteins/metabolism , Pore Forming Cytotoxic Proteins/genetics , Pore Forming Cytotoxic Proteins/metabolism , Pyroptosis , SARS-CoV-2/enzymology , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , SARS-CoV-2/pathogenicity
7.
Science ; 374(6571): eabh2444, 2021 Nov 26.
Article in English | MEDLINE | ID: mdl-34822296

ABSTRACT

Immune and tissue stem cells retain an epigenetic memory of inflammation that intensifies sensitivity to future encounters. We investigated whether and to what consequence stem cells possess and accumulate memories of diverse experiences. Monitoring a choreographed response to wounds, we found that as hair follicle stem cells leave their niche, migrate to repair damaged epidermis, and take up long-term foreign residence there, they accumulate long-lasting epigenetic memories of each experience, culminating in post-repair epigenetic adaptations that sustain the epidermal transcriptional program and surface barrier. Each memory is distinct, separable, and has its own physiological impact, collectively endowing these stem cells with heightened regenerative ability to heal wounds and broadening their tissue-regenerating tasks relative to their naïve counterparts.


Subject(s)
Epidermal Cells/cytology , Epigenesis, Genetic , Hair Follicle/cytology , Stem Cells/physiology , Adaptation, Physiological , Animals , Cell Movement , Chromatin/metabolism , Epidermal Cells/physiology , Homeostasis , Inflammation , Mice , Regeneration , Stem Cell Niche , Transcriptome , Wound Healing
8.
Nat Commun ; 12(1): 188, 2021 01 08.
Article in English | MEDLINE | ID: mdl-33420028

ABSTRACT

Nod-like receptor (NLR) proteins activate pyroptotic cell death and IL-1 driven inflammation by assembling and activating the inflammasome complex. Closely related sensor proteins NLRP1 and CARD8 undergo unique auto-proteolysis-dependent activation and are implicated in auto-inflammatory diseases; however, their mechanisms of activation are not understood. Here we report the structural basis of how the activating domains (FIINDUPA-CARD) of NLRP1 and CARD8 self-oligomerize to assemble distinct inflammasome complexes. Recombinant FIINDUPA-CARD of NLRP1 forms a two-layered filament, with an inner core of oligomerized CARD surrounded by an outer ring of FIINDUPA. Biochemically, self-assembled NLRP1-CARD filaments are sufficient to drive ASC speck formation in cultured human cells-a process that is greatly enhanced by NLRP1-FIINDUPA which forms oligomers in vitro. The cryo-EM structures of NLRP1-CARD and CARD8-CARD filaments, solved here at 3.7 Å, uncover unique structural features that enable NLRP1 and CARD8 to discriminate between ASC and pro-caspase-1. In summary, our findings provide structural insight into the mechanisms of activation for human NLRP1 and CARD8 and reveal how highly specific signaling can be achieved by heterotypic CARD interactions within the inflammasome complexes.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , CARD Signaling Adaptor Proteins/metabolism , Inflammasomes/chemistry , Inflammasomes/metabolism , Neoplasm Proteins/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/genetics , CARD Signaling Adaptor Proteins/genetics , Caspase 1/metabolism , Cryoelectron Microscopy , HEK293 Cells , Humans , Inflammasomes/genetics , Inflammation , Molecular Docking Simulation , Mutation , NLR Proteins , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Signal Transduction
9.
Science ; 370(6521)2020 12 04.
Article in English | MEDLINE | ID: mdl-33093214

ABSTRACT

Immune sensor proteins are critical to the function of the human innate immune system. The full repertoire of cognate triggers for human immune sensors is not fully understood. Here, we report that human NACHT, LRR, and PYD domains-containing protein 1 (NLRP1) is activated by 3C proteases (3Cpros) of enteroviruses, such as human rhinovirus (HRV). 3Cpros directly cleave human NLRP1 at a single site between Glu130 and Gly131 This cleavage triggers N-glycine-mediated degradation of the autoinhibitory NLRP1 N-terminal fragment via the cullinZER1/ZYG11B complex, which liberates the activating C-terminal fragment. Infection of primary human airway epithelial cells by live human HRV triggers NLRP1-dependent inflammasome activation and interleukin-18 secretion. Our findings establish 3Cpros as a pathogen-derived trigger for the human NLRP1 inflammasome and suggest that NLRP1 may contribute to inflammatory diseases of the airway.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Cysteine Endopeptidases/metabolism , Immunity, Innate , Inflammasomes/metabolism , Respiratory Mucosa/virology , Rhinovirus/enzymology , Viral Proteins/metabolism , 3C Viral Proteases , Adaptor Proteins, Signal Transducing/chemistry , Apoptosis Regulatory Proteins/chemistry , Cell Cycle Proteins/metabolism , Cullin Proteins/metabolism , Glutamine/chemistry , Glutamine/metabolism , Glycine/chemistry , Glycine/metabolism , HEK293 Cells , HeLa Cells , Humans , Interleukin-18/metabolism , NLR Proteins , Proteolysis
10.
J Mol Cell Cardiol ; 129: 118-129, 2019 04.
Article in English | MEDLINE | ID: mdl-30771309

ABSTRACT

Reactive oxygen species (ROS) play a key role in development of heart failure but, at a cellular level, their effects range from cytoprotection to induction of cell death. Understanding how this is regulated is crucial to develop novel strategies to ameliorate only the detrimental effects. Here, we revisited the fundamental hypothesis that the level of ROS per se is a key factor in the cellular response by applying different concentrations of H2O2 to cardiomyocytes. High concentrations rapidly reduced intracellular ATP and inhibited protein synthesis. This was associated with activation of AMPK which phosphorylated and inhibited Raptor, a crucial component of mTOR complex-1 that regulates protein synthesis. Inhibition of protein synthesis by high concentrations of H2O2 prevents synthesis of immediate early gene products required for downstream gene expression, and such mRNAs (many encoding proteins required to deal with oxidant stress) were only induced by lower concentrations. Lower concentrations of H2O2 promoted mTOR phosphorylation, associated with differential recruitment of some mRNAs to the polysomes for translation. Some of the upregulated genes induced by low H2O2 levels are cytoprotective. We identified p21Cip1/WAF1 as one such protein, and preventing its upregulation enhanced the rate of cardiomyocyte apoptosis. The data support the concept of a "redox rheostat" in which different degrees of ROS influence cell energetics and intracellular signalling pathways to regulate mRNA and protein expression. This sliding scale determines cell fate, modulating survival vs death.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Apoptosis , Gene Expression Regulation , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Adenosine Triphosphate/metabolism , Animals , Animals, Newborn , Apoptosis/drug effects , Cell Survival/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cytoprotection/drug effects , Doxorubicin/pharmacology , Enzyme Activation/drug effects , Genes, Immediate-Early , Hydrogen Peroxide/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Myocytes, Cardiac/drug effects , Oxidation-Reduction , Phosphorylation/drug effects , Polyribosomes/metabolism , Protein Biosynthesis/drug effects , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , Stress, Physiological/drug effects
11.
Elife ; 72018 12 06.
Article in English | MEDLINE | ID: mdl-30520726

ABSTRACT

Adult stem cells are responsible for life-long tissue maintenance. They reside in and interact with specialized tissue microenvironments (niches). Using murine hair follicle as a model, we show that when junctional perturbations in the niche disrupt barrier function, adjacent stem cells dramatically change their transcriptome independent of bacterial invasion and become capable of directly signaling to and recruiting immune cells. Additionally, these stem cells elevate cell cycle transcripts which reduce their quiescence threshold, enabling them to selectively proliferate within this microenvironment of immune distress cues. However, rather than mobilizing to fuel new tissue regeneration, these ectopically proliferative stem cells remain within their niche to contain the breach. Together, our findings expose a potential communication relay system that operates from the niche to the stem cells to the immune system and back. The repurposing of proliferation by these stem cells patch the breached barrier, stoke the immune response and restore niche integrity.


Subject(s)
Cell Proliferation/genetics , Gene Expression Profiling/methods , Hair Follicle/metabolism , Stem Cell Niche , Stem Cells/metabolism , Animals , Cell Communication/genetics , Cell Cycle/genetics , Cells, Cultured , Hair Follicle/cytology , Hair Follicle/ultrastructure , Homeostasis/genetics , Humans , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Electron, Transmission , Stem Cells/cytology , Stem Cells/ultrastructure
12.
Proc Natl Acad Sci U S A ; 113(11): E1506-15, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26912458

ABSTRACT

Adult tissue stem cells (SCs) reside in niches, which orchestrate SC behavior. SCs are typically used sparingly and exist in quiescence unless activated for tissue growth. Whether parsimonious SC use is essential to conserve long-term tissue-regenerating potential during normal homeostasis remains poorly understood. Here, we examine this issue by conditionally ablating a key transcription factor Forkhead box C1 (FOXC1) expressed in hair follicle SCs (HFSCs). FOXC1-deficient HFSCs spend less time in quiescence, leading to markedly shortened resting periods between hair cycles. The enhanced hair cycling accelerates HFSC expenditure, and impacts hair regeneration in aging mice. Interestingly, although FOXC1-deficient HFs can still form a new bulge that houses HFSCs for the next hair cycle, the older bulge is left unanchored. As the new hair emerges, the entire old bulge, including its reserve HFSCs and SC-inhibitory inner cell layer, is lost. We trace this mechanism first, to a marked increase in cell cycle-associated transcripts upon Foxc1 ablation, and second, to a downstream reduction in E-cadherin-mediated inter-SC adhesion. Finally, we show that when the old bulge is lost with each hair cycle, overall levels of SC-inhibitory factors are reduced, further lowering the threshold for HFSC activity. Taken together, our findings suggest that HFSCs have restricted potential in vivo, which they conserve by coupling quiescence to adhesion-mediated niche maintenance, thereby achieving long-term tissue homeostasis.


Subject(s)
Adult Stem Cells/physiology , Forkhead Transcription Factors/metabolism , Hair Follicle/cytology , Hair Follicle/physiology , Adult Stem Cells/metabolism , Aging , Animals , Cadherins/metabolism , Cell Adhesion/physiology , Cell Proliferation , Forkhead Transcription Factors/genetics , Gene Expression Regulation , Mice, Knockout , Mice, Mutant Strains , Regeneration , Stem Cell Niche
13.
Nat Cell Biol ; 16(2): 179-90, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24463605

ABSTRACT

Hair follicle stem cells (HFSCs) regenerate hair in response to Wnt signalling. Here, we unfold genome-wide transcriptional and chromatin landscapes of ß-catenin-TCF3/4-TLE circuitry, and genetically dissect their biological roles within the native HFSC niche. We show that during HFSC quiescence, TCF3, TCF4 and TLE (Groucho) bind coordinately and transcriptionally repress Wnt target genes. We also show that ß-catenin is dispensable for HFSC viability, and if TCF3/4 levels are sufficiently reduced, it is dispensable for proliferation. However, ß-catenin is essential to activate genes that launch hair follicle fate and suppress sebocyte fate determination. TCF3/4 deficiency mimics Wnt-ß-catenin-dependent activation of these hair follicle fate targets; TCF3 overexpression parallels their TLE4-dependent suppression. Our studies unveil TCF3/4-TLE histone deacetylases as a repressive rheostat, whose action can be relieved by Wnt-ß-catenin signalling. When TCF3/4 and TLE levels are high, HFSCs can maintain stemness, but remain quiescent. When these levels drop or when Wnt-ß-catenin levels rise, this balance is shifted and hair regeneration initiates.


Subject(s)
Hair Follicle/metabolism , Transcription, Genetic , Wnt Proteins/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Hair Follicle/cytology , Mice , Mice, Knockout , Regeneration , Signal Transduction , Transcription Factor 4 , beta Catenin/physiology
14.
Nat Immunol ; 14(7): 749-55, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23708250

ABSTRACT

Beyond its well-characterized functions in antibody diversification, the cytidine deaminase AID can catalyze off-target DNA damage and has been hypothesized to edit RNA and mediate DNA demethylation. To comprehensively examine the effects of AID on the transcriptome and the pattern of DNA methylation ('methylome'), we analyzed AID-deficient (Aicda(-/-)), wild-type and AID-overexpressing activated B cells by high-throughput RNA sequencing (RNA-Seq) and reduced-representation bisulfite sequencing (RRBS). These analyses confirmed the known role of AID in immunoglobulin isotype switching and also demonstrated few other effects of AID on gene expression. Additionally, we detected no evidence of AID-dependent editing of mRNA or microRNA. Finally, the RRBS data did not support the proposed role for AID in regulating DNA methylation. Thus, despite evidence of its additional activities in other systems, antibody diversification seems to be the sole physiological function of AID in activated B cells.


Subject(s)
B-Lymphocytes/immunology , Cytidine Deaminase/immunology , Immunoglobulin Isotypes/immunology , Somatic Hypermutation, Immunoglobulin/immunology , Transcriptome/immunology , Animals , Female , Male , Mice , Mice, Knockout , MicroRNAs/chemistry , MicroRNAs/genetics , RNA, Messenger/chemistry , RNA, Messenger/genetics , Sequence Analysis, DNA
15.
Hum Mol Genet ; 22(7): 1395-403, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23300193

ABSTRACT

In this paper, we report a novel heterozygous mutation of A285V codon conversion on exon 4 of the desmin (DES), using whole exome sequencing (WES) in an isolated proband with documented dilated cardiomyopathy (DCM). This mutation is predicted to cause three-dimensional structure changes of DES. Immunohistological and electron microscopy studies demonstrated diffuse abnormal DES aggregations in DCM-induced-pluripotent stem cell (iPSC)-derived cardiomyocytes, and control-iPSC-derived cardiomyocytes transduced with A285V-DES. DCM-iPSC-derived cardiomyocytes also exhibited functional abnormalities in vitro. This is the first demonstration that patient-specific iPSC-derived cardiomyocytes can be used to provide histological and functional confirmation of a suspected genetic basis for DCM identified by WES.


Subject(s)
Cardiomyopathy, Dilated/genetics , Desmin/genetics , Induced Pluripotent Stem Cells/physiology , Myocytes, Cardiac/metabolism , Adult , Amino Acid Sequence , Base Sequence , Cardiomyopathy, Dilated/diagnostic imaging , Cardiomyopathy, Dilated/physiopathology , Cell Differentiation , Desmin/chemistry , Desmin/metabolism , Exome , Exons , HEK293 Cells , High-Throughput Nucleotide Sequencing , Humans , Male , Molecular Sequence Data , Mutation, Missense , Pedigree , Phenotype , Sequence Analysis, DNA , Stroke Volume/genetics , Ultrasonography , Ventricular Dysfunction, Left/diagnostic imaging , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/physiopathology
16.
Elife ; 2: e00178, 2013 Jan 22.
Article in English | MEDLINE | ID: mdl-23359859

ABSTRACT

The neuronal RNA binding protein NOVA regulates splicing, shuttles to the cytoplasm, and co-localizes with target transcripts in dendrites, suggesting links between splicing and local translation. Here we identified >200 transcripts showing NOVA-dependent changes in abundance, but, surprisingly, HITS-CLIP revealed NOVA binds these RNAs in introns rather than 3' UTRs. This led us to discover NOVA-regulated splicing of cryptic exons within these introns. These exons triggered nonsense mediated decay (NMD), as UPF1 and protein synthesis were required for NOVA's effect on RNA levels. Their regulation was dynamic and physiologically relevant. The NMD exons were regulated by seizures, which also induced changes in Nova subcellular localization and mediated large changes in synaptic proteins, including proteins implicated in familial epilepsy. Moreover, Nova haploinsufficient mice had spontaneous epilepsy. The data reveal a hidden means of dynamic RNA regulation linking electrical activity to splicing and protein output, and of mediating homeostatic excitation/inhibition balance in neurons.DOI:http://dx.doi.org/10.7554/eLife.00178.001.


Subject(s)
Antigens, Neoplasm/physiology , Exons , Nerve Tissue Proteins/metabolism , RNA-Binding Proteins/physiology , Seizures/metabolism , Synapses/metabolism , 3' Untranslated Regions , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Brain/metabolism , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Neuro-Oncological Ventral Antigen , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Subcellular Fractions
17.
Stem Cells Transl Med ; 1(1): 70-8, 2012 Jan.
Article in English | MEDLINE | ID: mdl-23197643

ABSTRACT

Stem cells have considerable potential to repair damaged organs and tissues. We previously showed that prenatal transplantation of human first trimester fetal blood mesenchymal stem cells (hfMSCs) in a mouse model of osteogenesis imperfecta (oim mice) led to a phenotypic improvement, with a marked decrease in fracture rate. Donor cells differentiated into mature osteoblasts, producing bone proteins and minerals, including collagen type Iα2, which is absent in nontransplanted mice. This led to modifications of the bone matrix and subsequent decrease of bone brittleness, indicating that grafted cells directly contribute to improvement of bone mechanical properties. Nevertheless, the therapeutic effect was incomplete, attributing to the limited level of engraftment in bone. In this study, we show that although migration of hfMSCs to bone and bone marrow is CXCR4-SDF1 (SDF1 is stromal-derived factor) dependent, only a small number of cells present CXCR4 on the cell surface despite high levels of internal CXCR4. Priming with SDF1, however, upregulates CXCR4 to increase the CXCR4(+) cell fraction, improving chemotaxis in vitro and enhancing engraftment in vivo at least threefold in both oim and wild-type bone and bone marrow. Higher engraftment in oim bones was associated with decreased bone brittleness. This strategy represents a step to improve the therapeutic benefits of fetal cell therapy toward being curative.


Subject(s)
Fractures, Bone/prevention & control , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/immunology , Osteoblasts/transplantation , Osteogenesis Imperfecta/surgery , Receptors, CXCR4/metabolism , Animals , Bone Matrix/metabolism , Cell Differentiation , Cells, Cultured , Chemokine CXCL12/metabolism , Chemotaxis , Collagen Type I/deficiency , Collagen Type I/genetics , Collagen Type I/metabolism , Disease Models, Animal , Female , Fractures, Bone/metabolism , Fractures, Bone/pathology , Humans , Male , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoblasts/immunology , Osteoblasts/metabolism , Osteoblasts/pathology , Osteogenesis Imperfecta/genetics , Osteogenesis Imperfecta/immunology , Osteogenesis Imperfecta/metabolism , Osteogenesis Imperfecta/pathology , Time Factors , Up-Regulation
18.
Mol Ther ; 20(10): 1953-67, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22760542

ABSTRACT

Induced pluripotent stem cells (iPSCs) with potential for therapeutic applications can be derived from somatic cells via ectopic expression of a set of limited and defined transcription factors. However, due to risks of random integration of the reprogramming transgenes into the host genome, the low efficiency of the process, and the potential risk of virally induced tumorigenicity, alternative methods have been developed to generate pluripotent cells using nonintegrating systems, albeit with limited success. Here, we show that c-KIT+ human first-trimester amniotic fluid stem cells (AFSCs) can be fully reprogrammed to pluripotency without ectopic factors, by culture on Matrigel in human embryonic stem cell (hESC) medium supplemented with the histone deacetylase inhibitor (HDACi) valproic acid (VPA). The cells share 82% transcriptome identity with hESCs and are capable of forming embryoid bodies (EBs) in vitro and teratomas in vivo. After long-term expansion, they maintain genetic stability, protein level expression of key pluripotency factors, high cell-division kinetics, telomerase activity, repression of X-inactivation, and capacity to differentiate into lineages of the three germ layers, such as definitive endoderm, hepatocytes, bone, fat, cartilage, neurons, and oligodendrocytes. We conclude that AFSC can be utilized for cell banking of patient-specific pluripotent cells for potential applications in allogeneic cellular replacement therapies, pharmaceutical screening, and disease modeling.


Subject(s)
Amniotic Fluid/drug effects , Histone Deacetylase Inhibitors/pharmacology , Induced Pluripotent Stem Cells/drug effects , Valproic Acid/pharmacology , Amniotic Fluid/cytology , Cell Differentiation , Cell Line , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Genome, Human , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Karyotyping , Kinetics , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Male , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Phenotype , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Sequence Analysis, DNA , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptome , Transgenes , X Chromosome Inactivation/drug effects
19.
Aging (Albany NY) ; 4(11): 803-822, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23362510

ABSTRACT

AIMS: We identified an autosomal dominant non­sense mutation (R225X) in exon 4 of the lamin A/C (LMNA) gene in a Chinese family spanning 3 generations with familial dilated cardiomyopathy (DCM). In present study, we aim to generate induced pluripotent stem cells derived cardiomyocytes (iPSC­CMs) from an affected patient with R225X and another patient bearing LMNA frame­shift mutation for drug screening. METHODS AND RESULTS: Higher prevalence of nuclear bleb formation and micronucleation was present in LMNA(R225X/WT) and LMNA(Framshift/WT) iPSC­CMs. Under field electrical stimulation, percentage of LMNA­mutated iPSC­CMs exhibiting nuclear senescence and cellular apoptosis markedly increased. shRNA knockdown of LMNA replicated those phenotypes of the mutated LMNA field electrical stress. Pharmacological blockade of ERK1/2 pathway with MEK1/2 inhibitors, U0126 and selumetinib (AZD6244) significantly attenuated the pro­apoptotic effects of field electric stimulation on the mutated LMNA iPSC­CMs. CONCLUSION: LMNA­related DCM was modeled in­vitro using patient­specific iPSC­CMs. Our results demonstrated that haploinsufficiency due to R225X LMNA non­sense mutation was associated with accelerated nuclear senescence and apoptosis of iPSC­ CMs under electrical stimulation, which can be significantly attenuated by therapeutic blockade of stress­related ERK1/2 pathway.


Subject(s)
Aging/physiology , Cardiomyopathy, Dilated , Induced Pluripotent Stem Cells/cytology , Lamin Type A/genetics , Models, Biological , Myocytes, Cardiac/cytology , Blotting, Western , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Cell Differentiation/physiology , Female , Fibroblasts/cytology , Fluorescent Antibody Technique , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Middle Aged , Mutation , Myocytes, Cardiac/metabolism , Patch-Clamp Techniques , Pedigree
20.
J Biol Chem ; 284(40): 27195-210, 2009 Oct 02.
Article in English | MEDLINE | ID: mdl-19638633

ABSTRACT

The Mdm2 ubiquitin ligase is an important regulator of p53 abundance and p53-dependent apoptosis. Mdm2 expression is frequently regulated by a p53 Mdm2 autoregulatory loop whereby p53 stimulates Mdm2 expression and hence its own degradation. Although extensively studied in cell lines, relatively little is known about Mdm2 expression in heart where oxidative stress (exacerbated during ischemia-reperfusion) is an important pro-apoptotic stimulus. We demonstrate that Mdm2 transcript and protein expression are induced by oxidative stress (0.2 mm H(2)O(2)) in neonatal rat cardiac myocytes. In other cells, constitutive Mdm2 expression is regulated by the P1 promoter (5' to exon 1), with inducible expression regulated by the P2 promoter (in intron 1). In myocytes, H(2)O(2) increased Mdm2 expression from the P2 promoter, which contains two p53-response elements (REs), one AP-1 RE, and two Ets REs. H(2)O(2) did not detectably increase expression of p53 mRNA or protein but did increase expression of several AP-1 transcription factors. H(2)O(2) increased binding of AP-1 proteins (c-Jun, JunB, JunD, c-Fos, FosB, and Fra-1) to an Mdm2 AP-1 oligodeoxynucleotide probe, and chromatin immunoprecipitation assays showed it increased binding of c-Jun or JunB to the P2 AP-1 RE. Finally, antisense oligonucleotide-mediated reduction of H(2)O(2)-induced Mdm2 expression increased caspase 3 activation. Thus, increased Mdm2 expression is associated with transactivation at the P2 AP-1 RE (rather than the p53 or Ets REs), and Mdm2 induction potentially represents a cardioprotective response to oxidative stress.


Subject(s)
Gene Expression Regulation/drug effects , Hydrogen Peroxide/pharmacology , Myocytes, Cardiac/drug effects , Oxidative Stress/drug effects , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Sequence Homology, Amino Acid , Animals , Animals, Newborn , Apoptosis/drug effects , Apoptosis/genetics , Base Sequence , Caspase 3/metabolism , Humans , Introns/genetics , Mice , Molecular Sequence Data , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Oxidative Stress/genetics , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Response Elements , Transcription Factor AP-1/genetics , Transcriptional Activation/drug effects , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL