Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Commun Biol ; 7(1): 324, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38485785

ABSTRACT

Typical multiomics studies employ separate methods for DNA, RNA, and protein sample preparation, which is labor intensive, costly, and prone to sampling bias. We describe a method for preparing high-quality, sequencing-ready DNA and RNA, and either intact proteins or mass-spectrometry-ready peptides for whole proteome analysis from a single sample. This method utilizes a reversible protein tagging scheme to covalently link all proteins in a lysate to a bead-based matrix and nucleic acid precipitation and selective solubilization to yield separate pools of protein and nucleic acids. We demonstrate the utility of this method to compare the genomes, transcriptomes, and proteomes of four triple-negative breast cancer cell lines with different degrees of malignancy. These data show the involvement of both RNA and associated proteins, and protein-only dependent pathways that distinguish these cell lines. We also demonstrate the utility of this multiomics workflow for tissue analysis using mouse brain, liver, and lung tissue.


Subject(s)
Multiomics , RNA , Animals , Mice , DNA/genetics , Mass Spectrometry/methods , Proteome/metabolism , RNA/genetics
2.
Sci Rep ; 11(1): 18916, 2021 09 23.
Article in English | MEDLINE | ID: mdl-34556702

ABSTRACT

Amyotrophic lateral sclerosis is an adult-onset neurodegenerative disorder characterized by loss of motor neurons. Mitochondria are essential for neuronal survival but the developmental timing and mechanistic importance of mitochondrial dysfunction in sporadic ALS (sALS) neurons is not fully understood. We used human induced pluripotent stem cells and generated a developmental timeline by differentiating sALS iPSCs to neural progenitors and to motor neurons and comparing mitochondrial parameters with familial ALS (fALS) and control cells at each developmental stage. We report that sALS and fALS motor neurons have elevated reactive oxygen species levels, depolarized mitochondria, impaired oxidative phosphorylation, ATP loss and defective mitochondrial protein import compared with control motor neurons. This phenotype develops with differentiation into motor neurons, the affected cell type in ALS, and does not occur in the parental undifferentiated sALS cells or sALS neural progenitors. Our work demonstrates a developmentally regulated unifying mitochondrial phenotype between patient derived sALS and fALS motor neurons. The occurrence of a unifying mitochondrial phenotype suggests that mitochondrial etiology known to SOD1-fALS may applicable to sALS. Furthermore, our findings suggest that disease-modifying treatments focused on rescue of mitochondrial function may benefit both sALS and fALS patients.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Cell Differentiation , Mitochondria/pathology , Motor Neurons/pathology , Neural Stem Cells/pathology , Biopsy , Cells, Cultured , Fibroblasts , Humans , Induced Pluripotent Stem Cells , Motor Neurons/cytology , Motor Neurons/metabolism , Neural Stem Cells/cytology , Primary Cell Culture , Reactive Oxygen Species/metabolism , Skin/pathology
4.
J Clin Invest ; 130(6): 3124-3136, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32182222

ABSTRACT

Chronic inflammation is a pathologic feature of neurodegeneration and aging; however, the mechanism regulating this process is not understood. Melatonin, an endogenous free radical scavenger synthesized by neuronal mitochondria, decreases with aging and neurodegeneration. We proposed that insufficient melatonin levels impair mitochondrial homeostasis, resulting in mitochondrial DNA (mtDNA) release and activation of cytosolic DNA-mediated inflammatory response in neurons. We found increased mitochondrial oxidative stress and decreased mitochondrial membrane potential, with higher mtDNA release in brain and primary cerebro-cortical neurons of melatonin-deficient aralkylamine N-acetyltransferase (AANAT) knockout mice. Cytosolic mtDNA activated the cGAS/STING/IRF3 pathway, stimulating inflammatory cytokine generation. We found that Huntington's disease mice had increased mtDNA release, cGAS activation, and inflammation, all inhibited by exogenous melatonin. Thus, we demonstrated that cytosolic mtDNA activated the inflammatory response in aging and neurodegeneration, a process modulated by melatonin. Furthermore, our data suggest that AANAT knockout mice are a model of accelerated aging.


Subject(s)
Aging/metabolism , Cytosol/metabolism , DNA, Mitochondrial/metabolism , Huntington Disease/metabolism , Melatonin/pharmacology , Neurons/metabolism , Signal Transduction/drug effects , Aging/genetics , Aging/pathology , Animals , Cytosol/pathology , DNA, Mitochondrial/genetics , Female , Humans , Huntington Disease/genetics , Huntington Disease/pathology , Male , Mice , Mice, Knockout , Neurons/pathology
5.
J Huntingtons Dis ; 8(4): 449-457, 2019.
Article in English | MEDLINE | ID: mdl-31594239

ABSTRACT

BACKGROUND: Selective serotonin reuptake inhibitors (SSRIs) target the serotonin transporter (SERT) and are commonly prescribed for depression in Huntington's disease (HD) patients. However, SERT expression in HD has not been carefully evaluated in patients or mouse models. OBJECTIVE: In this study, we investigated SERT levels in HD patients and HD mouse models. METHODS: We obtained HD patient brain striatal samples and matched controls, as well as brain tissue from CAG140 and R6/2 mice. SERT mRNA and protein levels were analyzed using quantitative RT-PCR and immunoblotting. RESULTS AND CONCLUSIONS: We found that SERT protein, but not mRNA is markedly increased in grade 4 HD patient striatal tissue. These findings suggest posttranscriptional or translational SERT dysregulation as a possible etiologic factor modulating psychopathology in HD. Interestingly, SERT expression is variable in mouse models of the disease. Increased SERT levels are demonstrated in the brain of CAG140 mice, a full-length knock-in mouse model of the disease, but not in the striatum of the R6/2 fragment murine model of the disease. Based on this parameter, the CAG140 huntingtin knock-in mouse model is more suitable than the R6/2 model for the study of serotonergic pathway pathology in Huntington's disease.


Subject(s)
Corpus Striatum/metabolism , Huntington Disease/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Animals , Disease Models, Animal , Humans , Mice , Mice, Transgenic , RNA, Messenger/metabolism
6.
Proc Natl Acad Sci U S A ; 116(33): 16593-16602, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31346086

ABSTRACT

Mutant huntingtin (mHTT), the causative protein in Huntington's disease (HD), associates with the translocase of mitochondrial inner membrane 23 (TIM23) complex, resulting in inhibition of synaptic mitochondrial protein import first detected in presymptomatic HD mice. The early timing of this event suggests that it is a relevant and direct pathophysiologic consequence of mHTT expression. We show that, of the 4 TIM23 complex proteins, mHTT specifically binds to the TIM23 subunit and that full-length wild-type huntingtin (wtHTT) and mHTT reside in the mitochondrial intermembrane space. We investigated differences in mitochondrial proteome between wtHTT and mHTT cells and found numerous proteomic disparities between mHTT and wtHTT mitochondria. We validated these data by quantitative immunoblotting in striatal cell lines and human HD brain tissue. The level of soluble matrix mitochondrial proteins imported through the TIM23 complex is lower in mHTT-expressing cell lines and brain tissues of HD patients compared with controls. In mHTT-expressing cell lines, membrane-bound TIM23-imported proteins have lower intramitochondrial levels, whereas inner membrane multispan proteins that are imported via the TIM22 pathway and proteins integrated into the outer membrane generally remain unchanged. In summary, we show that, in mitochondria, huntingtin is located in the intermembrane space, that mHTT binds with high-affinity to TIM23, and that mitochondria from mHTT-expressing cells and brain tissues of HD patients have reduced levels of nuclearly encoded proteins imported through TIM23. These data demonstrate the mechanism and biological significance of mHTT-mediated inhibition of mitochondrial protein import, a mechanism likely broadly relevant to other neurodegenerative diseases.


Subject(s)
Huntingtin Protein/metabolism , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mutant Proteins/metabolism , Proteostasis , Cell Line , Cell Nucleus/metabolism , Cerebral Cortex/pathology , Corpus Striatum/pathology , Humans , Huntington Disease , Mitochondrial Membranes/metabolism , Mitochondrial Precursor Protein Import Complex Proteins , Mitochondrial Proteins/metabolism , Protein Binding , Proteome/metabolism
7.
Proc Natl Acad Sci U S A ; 116(2): 650-659, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30584104

ABSTRACT

Neuritic retraction in the absence of overt neuronal death is a shared feature of normal aging and neurodegenerative disorders, but the intracellular mechanisms modulating this process are not understood. We propose that cumulative distal mitochondrial protein damage results in impaired protein import, leading to mitochondrial dysfunction and focal activation of the canonical apoptosis pathway in neurites. This is a controlled process that may not lead to neuronal death and, thus, we term this phenomenon "neuritosis." Consistent with our hypothesis, we show that in primary cerebrocortical neurons, mitochondrial distance from the soma correlates with increased mitochondrial protein damage, PINK1 accumulation, reactive oxygen species production, and decreased mitochondrial membrane potential and depolarization threshold. Furthermore, we demonstrate that the distance-dependent mitochondrial membrane potential gradient exists in vivo in mice. We demonstrate that impaired distal mitochondria have a lower threshold for focal/nonlethal neuritic caspase-3 activation in normal neurons that is exacerbated in aging, stress, and neurodegenerative conditions, thus delineating a fundamental mechanistic underpinning for synaptic vulnerability.


Subject(s)
Apoptosis , Membrane Potential, Mitochondrial , Mitochondria/metabolism , Neurites/metabolism , Neurodegenerative Diseases/metabolism , Animals , Caspase 3/genetics , Caspase 3/metabolism , Mice , Mice, Transgenic , Mitochondria/genetics , Mitochondria/pathology , Neurites/pathology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Protein Kinases/genetics , Protein Kinases/metabolism , Reactive Oxygen Species/metabolism
8.
Proc Natl Acad Sci U S A ; 114(38): E7997-E8006, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28874589

ABSTRACT

G protein-coupled receptors (GPCRs) are classically characterized as cell-surface receptors transmitting extracellular signals into cells. Here we show that central components of a GPCR signaling system comprised of the melatonin type 1 receptor (MT1), its associated G protein, and ß-arrestins are on and within neuronal mitochondria. We discovered that the ligand melatonin is exclusively synthesized in the mitochondrial matrix and released by the organelle activating the mitochondrial MT1 signal-transduction pathway inhibiting stress-mediated cytochrome c release and caspase activation. These findings coupled with our observation that mitochondrial MT1 overexpression reduces ischemic brain injury in mice delineate a mitochondrial GPCR mechanism contributing to the neuroprotective action of melatonin. We propose a new term, "automitocrine," analogous to "autocrine" when a similar phenomenon occurs at the cellular level, to describe this unexpected intracellular organelle ligand-receptor pathway that opens a new research avenue investigating mitochondrial GPCR biology.


Subject(s)
Brain Injuries/metabolism , Brain Ischemia/metabolism , Melatonin/biosynthesis , Mitochondria/metabolism , Receptor, Melatonin, MT1/metabolism , Signal Transduction , Animals , Brain Injuries/genetics , Brain Ischemia/genetics , Cytochromes c/genetics , Cytochromes c/metabolism , Male , Melatonin/genetics , Mice , Mitochondria/genetics , Receptor, Melatonin, MT1/genetics
9.
J Neurosci Methods ; 263: 1-6, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26808294

ABSTRACT

BACKGROUND: Functional and structural properties of mitochondria are highly tissue and cell dependent, but isolation of highly purified human neuronal mitochondria is not currently available. NEW METHOD: We developed and validated a procedure to isolate purified neuronal mitochondria from brain tissue. The method combines Percoll gradient centrifugation to obtain synaptosomal fraction with nitrogen cavitation mediated synaptosome disruption and extraction of mitochondria using anti mitochondrial outer membrane protein antibodies conjugated to magnetic beads. The final products of isolation are non-synaptosomal mitochondria, which are a mixture of mitochondria isolated from different brain cells (i.e. neurons, astrocytes, oligodendrocytes, microglia) and synaptic mitochondria, which are of neuronal origin. This method is well suited for preparing functional mitochondria from human cortex tissue that is surgically extracted. RESULTS: The procedure produces mitochondria with minimal cytoplasmic contaminations that are functionally active based on measurements of mitochondrial respiration as well as mitochondrial protein import. The procedure requires approximately four hours for the isolation of human neuronal mitochondria and can also be used to isolate mitochondria from mouse/rat/monkey brains. COMPARISON WITH EXISTING METHODS AND CONCLUSIONS: This method will allow researchers to study highly enriched neuronal mitochondria without the confounding effect of cellular and organelle contaminants.


Subject(s)
Cerebral Cortex/cytology , Mitochondria/physiology , Neurons/ultrastructure , Antibodies/metabolism , Cell Fractionation , HLA Antigens/metabolism , Humans , Membrane Potential, Mitochondrial/physiology , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/immunology , Mitochondrial Precursor Protein Import Complex Proteins , Mitochondrial Proteins/metabolism , Synaptosomes/metabolism , Synaptosomes/ultrastructure
10.
Nat Neurosci ; 17(6): 822-31, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24836077

ABSTRACT

Mitochondrial dysfunction is associated with neuronal loss in Huntington's disease (HD), a neurodegenerative disease caused by an abnormal polyglutamine expansion in huntingtin (Htt). However, the mechanisms linking mutant Htt and mitochondrial dysfunction in HD remain unknown. We identify an interaction between mutant Htt and the TIM23 mitochondrial protein import complex. Remarkably, recombinant mutant Htt directly inhibited mitochondrial protein import in vitro. Furthermore, mitochondria from brain synaptosomes of presymptomatic HD model mice and from mutant Htt-expressing primary neurons exhibited a protein import defect, suggesting that deficient protein import is an early event in HD. The mutant Htt-induced mitochondrial import defect and subsequent neuronal death were attenuated by overexpression of TIM23 complex subunits, demonstrating that deficient mitochondrial protein import causes mutant Htt-induced neuronal death. Collectively, these findings provide evidence for a direct link between mutant Htt, mitochondrial dysfunction and neuronal pathology, with implications for mitochondrial protein import-based therapies in HD.


Subject(s)
Huntington Disease/genetics , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/genetics , Nerve Tissue Proteins/genetics , Aged , Animals , Cells, Cultured , Female , HEK293 Cells , Humans , Huntingtin Protein , Huntington Disease/pathology , Huntington Disease/therapy , Male , Mice , Mice, Inbred CBA , Mice, Transgenic , Middle Aged , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Proteins/metabolism , Mutation , Nerve Tissue Proteins/physiology , Protein Transport/genetics
11.
Microrna ; 2(1): 2-13, 2013.
Article in English | MEDLINE | ID: mdl-25070710

ABSTRACT

Human papillomaviruses (HPVs) are associated with the pathogenesis of a variety of human cancers, including cervical and oropharyngeal cancers. The HPV E6 and E7 oncogenes are usually expressed to high levels in these cancers. Previous studies have shown dysregulation of cellular microRNAs (miRNAs) in HPV-positive cell lines and cancer tissues and recent studies have identified a few miRNAs whose levels are altered in the presence of the viral E6 and E7 proteins. In order to identify all the cellular miRNAs whose expression may be affected by these oncoproteins, we carried out microarray analysis using human foreskin keratinocytes (HFKs) expressing either or both of these two proteins. These studies showed that 90 and 60 miRNAs were dysregulated in the presence of the E6 or the E7 protein, respectively. Of these, 43 miRNAs were similarly affected in HFK-E6 and/or HFK-E7 when compared to control cells. The joint expression of E6 and E7 proteins in HFKs caused changes in the levels of 64 miRNAs, of which 24 were similarly affected in HFK-E6 and/or HFK-E7 cells relative to controls. The microarray experiments were validated by quantitative real-time RT-PCR analysis of several differentially expressed miRNAs. Several miRNAs dysregulated by the E6 and/or E7 proteins are known to be altered in a variety of human cancers. Furthermore, previously known cellular targets of these miRNAs are involved in processes such as cell cycle regulation, apoptosis, cell-cell adhesion, cell mobility and proliferation, and alterations in their levels may contribute to HPV-associated carcinogenesis. Taken together, the results of our studies suggest that high risk HPV E6 and E7 proteins share the ability to regulate a subset of cellular miRNAs.


Subject(s)
Human papillomavirus 16/metabolism , Keratinocytes/metabolism , MicroRNAs/metabolism , Oncogene Proteins, Viral/metabolism , Papillomavirus E7 Proteins/metabolism , Repressor Proteins/metabolism , Cells, Cultured , Foreskin/cytology , Humans , Keratinocytes/cytology , Male , Oligonucleotide Array Sequence Analysis , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins/genetics , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/metabolism , Repressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...