Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 109
Filter
Add more filters

Publication year range
1.
Mol Cell ; 75(4): 835-848.e8, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31378462

ABSTRACT

Mitochondrial dysfunction and proteostasis failure frequently coexist as hallmarks of neurodegenerative disease. How these pathologies are related is not well understood. Here, we describe a phenomenon termed MISTERMINATE (mitochondrial-stress-induced translational termination impairment and protein carboxyl terminal extension), which mechanistically links mitochondrial dysfunction with proteostasis failure. We show that mitochondrial dysfunction impairs translational termination of nuclear-encoded mitochondrial mRNAs, including complex-I 30kD subunit (C-I30) mRNA, occurring on the mitochondrial surface in Drosophila and mammalian cells. Ribosomes stalled at the normal stop codon continue to add to the C terminus of C-I30 certain amino acids non-coded by mRNA template. C-terminally extended C-I30 is toxic when assembled into C-I and forms aggregates in the cytosol. Enhancing co-translational quality control prevents C-I30 C-terminal extension and rescues mitochondrial and neuromuscular degeneration in a Parkinson's disease model. These findings emphasize the importance of efficient translation termination and reveal unexpected link between mitochondrial health and proteome homeostasis mediated by MISTERMINATE.


Subject(s)
Codon, Terminator , Drosophila Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Mitochondrial Proteins/metabolism , Proteostasis Deficiencies/metabolism , Animals , Drosophila Proteins/genetics , Drosophila melanogaster , HeLa Cells , Humans , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Diseases/genetics , Mitochondrial Diseases/pathology , Mitochondrial Proteins/genetics , Proteostasis Deficiencies/genetics , Proteostasis Deficiencies/pathology , RNA, Mitochondrial/genetics , RNA, Mitochondrial/metabolism
2.
Dev Biol ; 507: 11-19, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38142805

ABSTRACT

Notch signaling controls numerous key cellular processes including cell fate determination and cell proliferation. Its malfunction has been linked to many developmental abnormalities and human disorders. Overactivation of Notch signaling is shown to be oncogenic. Retention of excess Notch protein in the endoplasmic reticulum (ER) can lead to altered Notch signaling and cell fate, but the mechanism is not well understood. In this study, we show that V5-tagged or untagged exogenous Notch is retained in the ER when overexpressed in fly tissues. Furthermore, we show that Notch retention in the ER leads to robust ER enlargement and elicits a rough eye phenotype. Gain-of-function of unfolded protein response (UPR) factors IRE1 or spliced Xbp1 (Xbp1-s) alleviates Notch accumulation in the ER, restores ER morphology and ameliorates the rough eye phenotype. Our results uncover a pivotal role of the IRE1/Xbp1 axis in regulating the detrimental effect of ER-localized excess Notch protein during development and tissue homeostasis.


Subject(s)
Drosophila Proteins , Drosophila , Animals , Humans , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress/genetics , Endoribonucleases/genetics , Endoribonucleases/metabolism , Homeostasis , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Unfolded Protein Response
3.
J Biol Chem ; 300(3): 105719, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38311171

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by dysregulation of the expression and processing of the amyloid precursor protein (APP). Protein quality control systems are dedicated to remove faulty and deleterious proteins to maintain cellular protein homeostasis (proteostasis). Identidying mechanisms underlying APP protein regulation is crucial for understanding AD pathogenesis. However, the factors and associated molecular mechanisms regulating APP protein quality control remain poorly defined. In this study, we show that mutant APP with its mitochondrial-targeting sequence ablated exhibited predominant endoplasmic reticulum (ER) distribution and led to aberrant ER morphology, deficits in locomotor activity, and shortened lifespan. We searched for regulators that could counteract the toxicity caused by the ectopic expression of this mutant APP. Genetic removal of the ribosome-associated quality control (RQC) factor RACK1 resulted in reduced levels of ectopically expressed mutant APP. By contrast, gain of RACK1 function increased mutant APP level. Additionally, overexpression of the ER stress regulator (IRE1) resulted in reduced levels of ectopically expressed mutant APP. Mechanistically, the RQC related ATPase VCP/p97 and the E3 ubiquitin ligase Hrd1 were required for the reduction of mutant APP level by IRE1. These factors also regulated the expression and toxicity of ectopically expressed wild type APP, supporting their relevance to APP biology. Our results reveal functions of RACK1 and IRE1 in regulating the quality control of APP homeostasis and mitigating its pathogenic effects, with implications for the understanding and treatment of AD.


Subject(s)
Alzheimer Disease , Amyloid beta-Protein Precursor , Drosophila Proteins , Endoribonucleases , Receptors for Activated C Kinase , Animals , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Protein Serine-Threonine Kinases , Receptors for Activated C Kinase/genetics , Receptors for Activated C Kinase/metabolism , Drosophila melanogaster , Disease Models, Animal , Endoribonucleases/genetics , Endoribonucleases/metabolism
4.
EMBO Rep ; 24(4): e55548, 2023 04 05.
Article in English | MEDLINE | ID: mdl-36794623

ABSTRACT

Mechanisms underlying the depletion of NAD+ and accumulation of reactive oxygen species (ROS) in aging and age-related disorders remain poorly defined. We show that reverse electron transfer (RET) at mitochondrial complex I, which causes increased ROS production and NAD+ to NADH conversion and thus lowered NAD+ /NADH ratio, is active during aging. Genetic or pharmacological inhibition of RET decreases ROS production and increases NAD+ /NADH ratio, extending the lifespan of normal flies. The lifespan-extending effect of RET inhibition is dependent on NAD+ -dependent Sirtuin, highlighting the importance of NAD+ /NADH rebalance, and on longevity-associated Foxo and autophagy pathways. RET and RET-induced ROS and NAD+ /NADH ratio changes are prominent in human induced pluripotent stem cell (iPSC) model and fly models of Alzheimer's disease (AD). Genetic or pharmacological inhibition of RET prevents the accumulation of faulty translation products resulting from inadequate ribosome-mediated quality control, rescues relevant disease phenotypes, and extends the lifespan of Drosophila and mouse AD models. Deregulated RET is therefore a conserved feature of aging, and inhibition of RET may open new therapeutic opportunities in the context of aging and age-related diseases including AD.


Subject(s)
Alzheimer Disease , Induced Pluripotent Stem Cells , Mice , Animals , Humans , NAD , Reactive Oxygen Species/metabolism , Electrons , Induced Pluripotent Stem Cells/metabolism , Aging/genetics , Aging/metabolism , Alzheimer Disease/genetics , Drosophila/genetics , Drosophila/metabolism
5.
Proc Natl Acad Sci U S A ; 119(42): e2202322119, 2022 10 18.
Article in English | MEDLINE | ID: mdl-36170200

ABSTRACT

An overarching goal of aging and age-related neurodegenerative disease research is to discover effective therapeutic strategies applicable to a broad spectrum of neurodegenerative diseases. Little is known about the extent to which targetable pathogenic mechanisms are shared among these seemingly diverse diseases. Translational control is critical for maintaining proteostasis during aging. Gaining control of the translation machinery is also crucial in the battle between viruses and their hosts. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the ongoing COVID-19 pandemic. Here, we show that overexpression of SARS-CoV-2-encoded nonstructural protein 1 (Nsp1) robustly rescued neuromuscular degeneration and behavioral phenotypes in Drosophila models of Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. These diseases share a common mechanism: the accumulation of aberrant protein species due to the stalling and collision of translating ribosomes, leading to proteostasis failure. Our genetic and biochemical analyses revealed that Nsp1 acted in a multipronged manner to resolve collided ribosomes, abort stalled translation, and remove faulty translation products causative of disease in these models, at least in part through the ribosome recycling factor ABCE1, ribosome-associated quality-control factors, autophagy, and AKT signaling. Nsp1 exhibited exquisite specificity in its action, as it did not modify other neurodegenerative conditions not known to be associated with ribosome stalling. These findings uncover a previously unrecognized mechanism of Nsp1 in manipulating host translation, which can be leveraged for combating age-related neurodegenerative diseases that are affecting millions of people worldwide and currently without effective treatment.


Subject(s)
COVID-19 , Neurodegenerative Diseases , RNA-Dependent RNA Polymerase , Ribosomes , Viral Nonstructural Proteins , Alzheimer Disease , Amyotrophic Lateral Sclerosis , Animals , COVID-19/genetics , Drosophila , Humans , Neurodegenerative Diseases/genetics , Pandemics , Parkinson Disease , Proto-Oncogene Proteins c-akt , RNA, Messenger/metabolism , Ribosomes/genetics , Ribosomes/metabolism , SARS-CoV-2/genetics , Viral Nonstructural Proteins/metabolism
6.
J Biol Chem ; 299(3): 102995, 2023 03.
Article in English | MEDLINE | ID: mdl-36764521

ABSTRACT

Expansion of G4C2 hexanucleotide repeats in the chromosome 9 ORF 72 (C9ORF72) gene is the most common genetic cause of amyotrophic lateral sclerosis (ALS) with frontotemporal dementia (C9-ALS/FTD). Dipeptide repeats generated by unconventional translation, especially the R-containing poly(GR), have been implicated in C9-ALS/FTD pathogenesis. Mutations in other genes, including TAR DNA-binding protein 43 KD (TDP-43), fused in sarcoma (FUS), and valosin-containing protein, have also been linked to ALS/FTD, and upregulation of amyloid precursor protein (APP) is observed at the early stage of ALS and FTD. Fundamental questions remain as to the relationships between these ALS/FTD genes and whether they converge on similar cellular pathways. Here, using biochemical, cell biological, and genetic analyses in Drosophila disease models, patient-derived fibroblasts, and mammalian cell culture, we show that mechanistic target of rapamycin complex 2 (mTORC2)/AKT signaling is activated by APP, TDP-43, and FUS and that mTORC2/AKT and its downstream target valosin-containing protein mediate the effect of APP, TDP-43, and FUS on the quality control of C9-ALS/FTD-associated poly(GR) translation. We also find that poly(GR) expression results in reduction of global translation and that the coexpression of APP, TDP-43, and FUS results in further reduction of global translation, presumably through the GCN2/eIF2α-integrated stress response pathway. Together, our results implicate mTORC2/AKT signaling and GCN2/eIF2α-integrated stress response as common signaling pathways underlying ALS/FTD pathogenesis.


Subject(s)
Amyotrophic Lateral Sclerosis , Frontotemporal Dementia , Animals , Amyotrophic Lateral Sclerosis/metabolism , C9orf72 Protein/genetics , C9orf72 Protein/metabolism , Dipeptides/metabolism , DNA Repeat Expansion , DNA-Binding Proteins/metabolism , Drosophila/metabolism , Frontotemporal Dementia/pathology , Mammals/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Quality Control , Valosin Containing Protein/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism
7.
Proc Natl Acad Sci U S A ; 117(40): 25104-25115, 2020 10 06.
Article in English | MEDLINE | ID: mdl-32958650

ABSTRACT

Maintaining the fidelity of nascent peptide chain (NP) synthesis is essential for proteome integrity and cellular health. Ribosome-associated quality control (RQC) serves to resolve stalled translation, during which untemplated Ala/Thr residues are added C terminally to stalled peptide, as shown during C-terminal Ala and Thr addition (CAT-tailing) in yeast. The mechanism and biological effects of CAT-tailing-like activity in metazoans remain unclear. Here we show that CAT-tailing-like modification of poly(GR), a dipeptide repeat derived from amyotrophic lateral sclerosis with frontotemporal dementia (ALS/FTD)-associated GGGGCC (G4C2) repeat expansion in C9ORF72, contributes to disease. We find that poly(GR) can act as a mitochondria-targeting signal, causing some poly(GR) to be cotranslationally imported into mitochondria. However, poly(GR) translation on mitochondrial surface is frequently stalled, triggering RQC and CAT-tailing-like C-terminal extension (CTE). CTE promotes poly(GR) stabilization, aggregation, and toxicity. Our genetic studies in Drosophila uncovered an important role of the mitochondrial protease YME1L in clearing poly(GR), revealing mitochondria as major sites of poly(GR) metabolism. Moreover, the mitochondria-associated noncanonical Notch signaling pathway impinges on the RQC machinery to restrain poly(GR) accumulation, at least in part through the AKT/VCP axis. The conserved actions of YME1L and noncanonical Notch signaling in animal models and patient cells support their fundamental involvement in ALS/FTD.


Subject(s)
ATPases Associated with Diverse Cellular Activities/genetics , Amyotrophic Lateral Sclerosis/genetics , C9orf72 Protein/genetics , Drosophila Proteins/genetics , Frontotemporal Dementia/genetics , Metalloendopeptidases/genetics , Mitochondrial Proteins/genetics , Proteome/genetics , Receptors, Notch/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Arginine/genetics , DNA Repeat Expansion/genetics , Disease Models, Animal , Drosophila melanogaster/genetics , Frontotemporal Dementia/metabolism , Frontotemporal Dementia/pathology , HEK293 Cells , Humans , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Protein Biosynthesis , Ribosomes/genetics , Ribosomes/metabolism , Signal Transduction/genetics
8.
Hum Mol Genet ; 29(4): 541-553, 2020 03 13.
Article in English | MEDLINE | ID: mdl-31628467

ABSTRACT

Missense mutations in the RNA exosome component exosome component 2 (EXOSC2), also known as ribosomal RNA-processing protein 4 (RRP4), were recently identified in two unrelated families with a novel syndrome known as Short stature, Hearing loss, Retinitis pigmentosa and distinctive Facies (SHRF, #OMIM 617763). Little is known about the mechanism of the SHRF pathogenesis. Here we have studied the effect of mutations in EXOSC2/RRP4 in patient-derived lymphoblasts, clustered regularly interspaced short palindromic repeats (CRISPR)-generated mutant fetal keratinocytes and Drosophila. We determined that human EXOSC2 is an essential gene and that the pathogenic G198D mutation prevents binding to other RNA exosome components, resulting in protein and complex instability and altered expression and/or activities of critical genes, including those in the autophagy pathway. In parallel, we generated multiple CRISPR knockouts of the fly rrp4 gene. Using these flies, as well as rrp4 mutants with Piggy Bac (PBac) transposon insertion in the 3'UTR and RNAi flies, we determined that fly rrp4 was also essential, that fly rrp4 phenotypes could be rescued by wild-type human EXOSC2 but not the pathogenic form and that fly rrp4 is critical for eye development and maintenance, muscle ultrastructure and wing vein development. We found that overexpression of the transcription factor MITF was sufficient to rescue the small eye and adult lethal phenotypes caused by rrp4 inhibition. The autophagy genes ATG1 and ATG17, which are regulated by MITF, had similar effect. Pharmacological stimulation of autophagy with rapamycin also rescued the lethality caused by rrp4 inactivation. Our results implicate defective autophagy in SHRF pathogenesis and suggest therapeutic strategies.


Subject(s)
Exosome Multienzyme Ribonuclease Complex/genetics , RNA-Binding Proteins/genetics , Animals , Autophagy/genetics , Disease Models, Animal , Drosophila/genetics , Dwarfism/genetics , Exosome Multienzyme Ribonuclease Complex/metabolism , Exosomes/metabolism , Female , Genomics/methods , HEK293 Cells , Hearing Loss/genetics , Humans , Male , Mutation, Missense/genetics , Phenotype , RNA/metabolism , RNA-Binding Proteins/metabolism , Retinitis Pigmentosa/genetics , Syndrome
9.
Sensors (Basel) ; 22(3)2022 Jan 21.
Article in English | MEDLINE | ID: mdl-35161547

ABSTRACT

Thin-film silicon (Si)-based transient electronics represents an emerging technology that enables spontaneous dissolution, absorption and, finally, physical disappearance in a controlled manner under physiological conditions, and has attracted increasing attention in pertinent clinical applications such as biomedical implants for on-body sensing, disease diagnostics, and therapeutics. The degradation behavior of thin-film Si materials and devices is critically dependent on the device structure as well as the environment. In this work, we experimentally investigated the dissolution of planar Si thin films and micropatterned Si pillar arrays in a cell culture medium, and systematically analyzed the evolution of their topographical, physical, and chemical properties during the hydrolysis. We discovered that the cell culture medium significantly accelerates the degradation process, and Si pillar arrays present more prominent degradation effects by creating rougher surfaces, complicating surface states, and decreasing the electrochemical impedance. Additionally, the dissolution process leads to greatly reduced mechanical strength. Finally, in vitro cell culture studies demonstrate desirable biocompatibility of corroded Si pillars. The results provide a guideline for the use of thin-film Si materials and devices as transient implants in biomedicine.


Subject(s)
Electronics , Silicon , Cell Culture Techniques , Indicators and Reagents
10.
Proc Natl Acad Sci U S A ; 115(38): E8844-E8853, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30185553

ABSTRACT

Calcium (Ca2+) homeostasis is essential for neuronal function and survival. Altered Ca2+ homeostasis has been consistently observed in neurological diseases. How Ca2+ homeostasis is achieved in various cellular compartments of disease-relevant cell types is not well understood. Here we show in Drosophila Parkinson's disease (PD) models that Ca2+ transport from the endoplasmic reticulum (ER) to mitochondria through the ER-mitochondria contact site (ERMCS) critically regulates mitochondrial Ca2+ (mito-Ca2+) homeostasis in dopaminergic (DA) neurons, and that the PD-associated PINK1 protein modulates this process. In PINK1 mutant DA neurons, the ERMCS is strengthened and mito-Ca2+ level is elevated, resulting in mitochondrial enlargement and neuronal death. Miro, a well-characterized component of the mitochondrial trafficking machinery, mediates the effects of PINK1 on mito-Ca2+ and mitochondrial morphology, apparently in a transport-independent manner. Miro overexpression mimics PINK1 loss-of-function effect, whereas inhibition of Miro or components of the ERMCS, or pharmacological modulation of ERMCS function, rescued PINK1 mutant phenotypes. Mito-Ca2+ homeostasis is also altered in the LRRK2-G2019S model of PD and the PAR-1/MARK model of neurodegeneration, and genetic or pharmacological restoration of mito-Ca2+ level is beneficial in these models. Our results highlight the importance of mito-Ca2+ homeostasis maintained by Miro and the ERMCS to mitochondrial physiology and neuronal integrity. Targeting this mito-Ca2+ homeostasis pathway holds promise for a therapeutic strategy for neurodegenerative diseases.


Subject(s)
Calcium/metabolism , Drosophila melanogaster/metabolism , Endoplasmic Reticulum/metabolism , Mitochondria/metabolism , Parkinson Disease/pathology , Animals , Animals, Genetically Modified , Chelating Agents/pharmacology , Disease Models, Animal , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Endoplasmic Reticulum/drug effects , Glycogen Synthase Kinase 3/metabolism , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Loss of Function Mutation , Mitochondria/drug effects , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , rho GTP-Binding Proteins/metabolism
11.
Genes Dev ; 27(24): 2642-7, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24352421

ABSTRACT

The self-renewal versus differentiation choice of Drosophila and mammalian neural stem cells (NSCs) requires Notch (N) signaling. How N regulates NSC behavior is not well understood. Here we show that canonical N signaling cooperates with a noncanonical N signaling pathway to mediate N-directed NSC regulation. In the noncanonical pathway, N interacts with PTEN-induced kinase 1 (PINK1) to influence mitochondrial function, activating mechanistic target of rapamycin complex 2 (mTORC2)/AKT signaling. Importantly, attenuating noncanonical N signaling preferentially impaired the maintenance of Drosophila and human cancer stem cell-like tumor-forming cells. Our results emphasize the importance of mitochondria to N and NSC biology, with important implications for diseases associated with aberrant N signaling.


Subject(s)
Mitochondria/metabolism , Multiprotein Complexes/metabolism , Neoplastic Stem Cells/metabolism , Protein Kinases/metabolism , Receptors, Notch/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Animals , Brain Neoplasms/physiopathology , Cell Line, Tumor , Cell Proliferation , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Gene Expression Regulation , Humans , Mechanistic Target of Rapamycin Complex 2 , Microscopy, Electron, Transmission , Mitochondria/enzymology , Mitochondria/ultrastructure , Multiprotein Complexes/genetics , Mutation , Protein Kinases/genetics , RNA Interference , TOR Serine-Threonine Kinases/genetics
12.
Genes Dev ; 27(2): 157-62, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23348839

ABSTRACT

Eukaryotes employ elaborate mitochondrial quality control (MQC) to maintain the function of the power-generating organelle. Parkinson's disease-associated PINK1 and Parkin actively participate in MQC. However, the signaling events involved are largely unknown. Here we show that mechanistic target of rapamycin 2 (mTORC2) and Tricornered (Trc) kinases act downstream from PINK1 to regulate MQC. Trc is phosphorylated in mTORC2-dependent and mTORC2-independent manners and is specifically localized to mitochondria in response to PINK1, which regulates mTORC2 through mitochondrial complex-I activity. Genetically, mTORC2 and Trc act upstream of Parkin. Thus, multiplex kinase signaling is acting between PINK1 and Parkin to regulate MQC, a process highly conserved in mammals.


Subject(s)
Drosophila Proteins/metabolism , Mitochondria/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Animals , Drosophila Proteins/genetics , Humans , Male , Protein Kinases/genetics , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/genetics
13.
PLoS Genet ; 13(5): e1006785, 2017 May.
Article in English | MEDLINE | ID: mdl-28520736

ABSTRACT

Notch (N) signaling is central to the self-renewal of neural stem cells (NSCs) and other tissue stem cells. Its deregulation compromises tissue homeostasis and contributes to tumorigenesis and other diseases. How N regulates stem cell behavior in health and disease is not well understood. Here we show that N regulates bantam (ban) microRNA to impact cell growth, a process key to NSC maintenance and particularly relied upon by tumor-forming cancer stem cells. Notch signaling directly regulates ban expression at the transcriptional level, and ban in turn feedback regulates N activity through negative regulation of the Notch inhibitor Numb. This feedback regulatory mechanism helps maintain the robustness of N signaling activity and NSC fate. Moreover, we show that a Numb-Myc axis mediates the effects of ban on nucleolar and cellular growth independently or downstream of N. Our results highlight intricate transcriptional as well as translational control mechanisms and feedback regulation in the N signaling network, with important implications for NSC biology and cancer biology.


Subject(s)
Brain/metabolism , Drosophila Proteins/genetics , Drosophila/genetics , Juvenile Hormones/genetics , MicroRNAs/genetics , Neoplastic Stem Cells/metabolism , Receptors, Notch/metabolism , Animals , Brain/cytology , Cell Differentiation , Cell Growth Processes , Drosophila/cytology , Drosophila/metabolism , Drosophila Proteins/metabolism , Feedback, Physiological , Juvenile Hormones/metabolism , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/physiology , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neural Stem Cells/physiology , Receptors, Notch/genetics , Signal Transduction
14.
Int J Hyperthermia ; 36(1): 632-639, 2019.
Article in English | MEDLINE | ID: mdl-31244349

ABSTRACT

Purpose: Microwave ablation (MWA) has become increasingly popular as a minimally invasive treatment for benign and malignant liver tumors. However, few studies have demonstrated the benefits and disadvantages of MWA compared to surgical resection (SR) for large hepatic hemangiomas. This study aimed to evaluate the safety and effectiveness of MWA compared to SR for large (5-10 cm) hepatic hemangiomas. Methods and materials: This retrospective comparative study included 112 patients with large, symptomatic hepatic hemangiomas who had been treated with MWA (n = 44) or SR (n = 68) and followed up for a median of 44 months using enhanced computed tomography (CT) or magnetic resonance imaging (MRI). Intraoperative information, postoperative recovery time, postoperative discomfort and complications and treatment effectiveness between groups were compared using a chi-square test or an independent t-test. Results: The operative time was significantly shorter (31.3 ± 21.76 versus 148.1 ± 59.3 min, p < .001) and the blood loss (10.2 ± 60.6 versus 227.9 ± 182.9 mL, p < .0001) and rate of prophylactic abdominal drainage [1 (2.3%) versus 57 (83.8%), p < .001] were significantly lower in the MWA group than in the SR group. Postoperative recovery of the MWA group in regard to indwelling catheter time, normal diet time, incision cicatrization time and hospital stay (p < .001) was significantly better than the SR group. However, no statistically significant difference in effectiveness was noted between the groups (p = .58). Conclusions: MWA may be as effective as SR, and potentially safer for treating large, symptomatic hepatic hemangiomas. To confirm our findings, large-sample, multicentered, randomized controlled trials are needed.


Subject(s)
Catheter Ablation/methods , Hemangioma/surgery , Liver Neoplasms/surgery , Microwaves/therapeutic use , Female , Hemangioma/diagnostic imaging , Humans , Liver Neoplasms/diagnostic imaging , Male , Middle Aged , Treatment Outcome , Ultrasonography
15.
Genes Dev ; 25(24): 2644-58, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22190460

ABSTRACT

Cancer stem cells (CSCs) are postulated to be a small subset of tumor cells with tumor-initiating ability that shares features with normal tissue-specific stem cells. The origin of CSCs and the mechanisms underlying their genesis are poorly understood, and it is uncertain whether it is possible to obliterate CSCs without inadvertently damaging normal stem cells. Here we show that a functional reduction of eukaryotic translation initiation factor 4E (eIF4E) in Drosophila specifically eliminates CSC-like cells in the brain and ovary without having discernable effects on normal stem cells. Brain CSC-like cells can arise from dedifferentiation of transit-amplifying progenitors upon Notch hyperactivation. eIF4E is up-regulated in these dedifferentiating progenitors, where it forms a feedback regulatory loop with the growth regulator dMyc to promote cell growth, particularly nucleolar growth, and subsequent ectopic neural stem cell (NSC) formation. Cell growth regulation is also a critical component of the mechanism by which Notch signaling regulates the self-renewal of normal NSCs. Our findings highlight the importance of Notch-regulated cell growth in stem cell maintenance and reveal a stronger dependence on eIF4E function and cell growth by CSCs, which might be exploited therapeutically.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/physiology , Receptors, Notch/metabolism , Animals , Brain/cytology , Brain/metabolism , Cell Differentiation , Cell Proliferation , Cell Size , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Gene Expression Regulation , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/metabolism , Neural Stem Cells/metabolism , Receptors, Notch/genetics , Signal Transduction
16.
Hum Mol Genet ; 25(23): 5059-5068, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27794540

ABSTRACT

Dysregulation of Fused in Sarcoma (FUS) gene expression is associated with fronto-temporal lobar degeneration (FTLD), and missense mutations in the FUS gene have been identified in patients affected by amyotrophic lateral sclerosis (ALS). However, molecular and cellular defects underlying FUS proteinopathy remain to be elucidated. Here, we examined whether genes important for mitochondrial quality control play a role in FUS proteinopathy. In our genetic screening, Pink1 and Park genes were identified as modifiers of neurodegeneration phenotypes induced by wild type (Wt) or ALS-associated P525L-mutant human FUS. Down-regulating expression of either Pink1 or Parkin genes ameliorated FUS-induced neurodegeneration phenotypes. The protein levels of PINK1 and Parkin were elevated in cells overexpressing FUS. Remarkably, ubiquitinylation of Miro1 protein, a downstream target of the E3 ligase activity of Parkin, was also increased in cells overexpressing FUS protein. In fly motor neurons expressing FUS, both motility and processivity of mitochondrial axonal transport were reduced by expression of either Wt- or P525L-mutant FUS. Finally, down-regulating PINK1 or Parkin partially rescued the locomotive defects and enhanced the survival rate in transgenic flies expressing FUS. Our data indicate that PINK1 and Parkin play an important role in FUS-induced neurodegeneration. This study has uncovered a previously unknown link between FUS proteinopathy and PINK1/Parkin genes, providing new insights into the pathogenesis of FUS proteinopathy.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Drosophila Proteins/genetics , Frontotemporal Lobar Degeneration/genetics , Heterogeneous-Nuclear Ribonucleoprotein Group F-H/genetics , Nerve Degeneration/genetics , Protein Serine-Threonine Kinases/genetics , Ubiquitin-Protein Ligases/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Animals, Genetically Modified , Axonal Transport/genetics , Disease Models, Animal , Frontotemporal Lobar Degeneration/physiopathology , Gene Expression Regulation , Genes, Modifier/genetics , Humans , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Motor Neurons/metabolism , Motor Neurons/pathology , Mutation, Missense , Nerve Degeneration/pathology , Phenotype , rho GTP-Binding Proteins/genetics
17.
Proc Natl Acad Sci U S A ; 111(5): 1927-32, 2014 Feb 04.
Article in English | MEDLINE | ID: mdl-24449853

ABSTRACT

Here, we report advanced materials and devices that enable high-efficiency mechanical-to-electrical energy conversion from the natural contractile and relaxation motions of the heart, lung, and diaphragm, demonstrated in several different animal models, each of which has organs with sizes that approach human scales. A cointegrated collection of such energy-harvesting elements with rectifiers and microbatteries provides an entire flexible system, capable of viable integration with the beating heart via medical sutures and operation with efficiencies of ∼2%. Additional experiments, computational models, and results in multilayer configurations capture the key behaviors, illuminate essential design aspects, and offer sufficient power outputs for operation of pacemakers, with or without battery assist.


Subject(s)
Diaphragm/physiology , Electric Power Supplies , Electrophysiological Phenomena , Heart/physiology , Lung/physiology , Motion , Animals , Cattle , Humans , Rats , Sheep
18.
Cell Biol Int ; 40(6): 671-85, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27005844

ABSTRACT

Bone marrow-derived mesenchymal stem cells (BM-MSCs) have been demonstrated as an ideal autologous stem cells source for cell-based therapy for myocardial infarction (MI). However, poor viability of donor stem cells after transplantation limits their therapeutic efficiency, whereas the underlying mechanism is still poorly understood. Autophagy, a highly conserved process of cellular degradation, is required for maintaining homeostasis and normal function. Here, we investigated the potential role of autophagy on apoptosis in BM-MSCs induced by hypoxic injury. BM-MSCs, isolated from male C57BL/6 mice, were subjected to hypoxia and serum deprivation (H/SD) injury for 6, 12, and 24 h, respectively. The autophagy state was regulated by 3-methyladenine (3MA) and rapamycin administration. Furthermore, compound C was administrated to inhibit AMPK. The apoptosis induced by H/SD was determined by TUNEL assays. Meanwhile, autophagy was measured by GFP-LC3 plasmids transfection and transmission electron microscope. Moreover, protein expressions were evaluated by Western blot assay. In the present study, we found that hypoxic stress increased autophagy and apoptosis in BM-MSCs time dependently. Meanwhile, hypoxia increased the activity of AMPK/mTOR signal pathway. Moreover, increased apoptosis in BM-MSCs under hypoxia was abolished by 3-MA, whereas was aggravated by rapamycin. Furthermore, the increased autophagy and apoptosis in BM-MSCs induced by hypoxia were abolished by AMPK inhibitor compound C. These data provide evidence that hypoxia induced AMPK/mTOR signal pathway activation which regulated the apoptosis and autophagy in BM-MSCs. Furthermore, the apoptosis of BM-MSCs under hypoxic condition was regulated by autophagy via AMPK/mTOR pathway.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Apoptosis/physiology , Mesenchymal Stem Cells/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Autophagy/physiology , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Hypoxia/physiology , Male , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred C57BL , Signal Transduction
19.
Nature ; 466(7306): 637-41, 2010 Jul 29.
Article in English | MEDLINE | ID: mdl-20671708

ABSTRACT

Gain-of-function mutations in leucine-rich repeat kinase 2 (LRRK2) cause familial as well as sporadic Parkinson's disease characterized by age-dependent degeneration of dopaminergic neurons. The molecular mechanism of LRRK2 action is not known. Here we show that LRRK2 interacts with the microRNA (miRNA) pathway to regulate protein synthesis. Drosophila e2f1 and dp messenger RNAs are translationally repressed by let-7 and miR-184*, respectively. Pathogenic LRRK2 antagonizes these miRNAs, leading to the overproduction of E2F1/DP, previously implicated in cell cycle and survival control and shown here to be critical for LRRK2 pathogenesis. Genetic deletion of let-7, antagomir-mediated blockage of let-7 and miR-184* action, transgenic expression of dp target protector, or replacement of endogenous dp with a dp transgene non-responsive to let-7 each had toxic effects similar to those of pathogenic LRRK2. Conversely, increasing the level of let-7 or miR-184* attenuated pathogenic LRRK2 effects. LRRK2 associated with Drosophila Argonaute-1 (dAgo1) or human Argonaute-2 (hAgo2) of the RNA-induced silencing complex (RISC). In aged fly brain, dAgo1 protein level was negatively regulated by LRRK2. Further, pathogenic LRRK2 promoted the association of phospho-4E-BP1 with hAgo2. Our results implicate deregulated synthesis of E2F1/DP caused by the miRNA pathway impairment as a key event in LRRK2 pathogenesis and suggest novel miRNA-based therapeutic strategies.


Subject(s)
Down-Regulation , MicroRNAs/genetics , MicroRNAs/metabolism , Protein Biosynthesis , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Animals , Argonaute Proteins , Cell Line , Dopamine/metabolism , Drosophila Proteins/biosynthesis , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster , E2F1 Transcription Factor/biosynthesis , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factors/biosynthesis , Eukaryotic Initiation Factors/metabolism , Female , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Male , MicroRNAs/antagonists & inhibitors , Neurons/cytology , Neurons/metabolism , Parkinson Disease/etiology , Parkinson Disease/genetics , Parkinson Disease/metabolism , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Induced Silencing Complex/antagonists & inhibitors , RNA-Induced Silencing Complex/chemistry , RNA-Induced Silencing Complex/metabolism , Trans-Activators/biosynthesis , Trans-Activators/genetics , Trans-Activators/metabolism , Up-Regulation
20.
J Cell Sci ; 126(Pt 3): 838-49, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23264732

ABSTRACT

Axon degeneration is observed at the early stages of many neurodegenerative conditions and this often leads to subsequent neuronal loss. We previously showed that inactivating the c-Jun N-terminal kinase (JNK) pathway leads to axon degeneration in Drosophila mushroom body (MB) neurons. To understand this process, we screened candidate suppressor genes and found that the Wallerian degeneration slow (Wld(S)) protein blocked JNK axonal degeneration. Although the nicotinamide mononucleotide adenylyltransferase (Nmnat1) portion of Wld(S) is required, we found that its nicotinamide adenine dinucleotide (NAD(+)) enzyme activity and the Wld(S) N-terminus (N70) are dispensable, unlike axotomy models of neurodegeneration. We suggest that Wld(S)-Nmnat protects against axonal degeneration through chaperone activity. Furthermore, ectopically expressed heat shock proteins (Hsp26 and Hsp70) also protected against JNK and Nmnat degeneration phenotypes. These results suggest that molecular chaperones are key in JNK- and Nmnat-regulated axonal protective functions.


Subject(s)
Axons/metabolism , Drosophila melanogaster/metabolism , Molecular Chaperones/metabolism , Nerve Tissue Proteins/metabolism , Wallerian Degeneration/metabolism , ADP Ribose Transferases/metabolism , Animals , Axons/pathology , Drosophila Proteins/metabolism , HSP72 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/metabolism , MAP Kinase Kinase 4/metabolism , Mushroom Bodies/pathology , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Signal Transduction , Wallerian Degeneration/pathology
SELECTION OF CITATIONS
SEARCH DETAIL