Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
FASEB J ; 38(4): e23494, 2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38376922

ABSTRACT

Pathological opening of the mitochondrial permeability transition pore (mPTP) is implicated in the pathogenesis of many disease processes such as myocardial ischemia, traumatic brain injury, Alzheimer's disease, and diabetes. While we have gained insight into mPTP biology over the last several decades, the lack of translation of this knowledge into successful clinical therapies underscores the need for continued investigation and use of different approaches to identify novel regulators of the mPTP with the hope of elucidating new therapeutic targets. Although the mPTP is known to be a voltage-gated channel, the identity of its voltage sensor remains unknown. Here we found decreased gating potential of the mPTP and increased expression and activity of sulfide quinone oxidoreductase (SQOR) in newborn Fragile X syndrome (FXS) mouse heart mitochondria, a model system of coenzyme Q excess and relatively decreased mPTP open probability. We further found that pharmacological inhibition and genetic silencing of SQOR increased mPTP open probability in vitro in adult murine cardiac mitochondria and in the isolated-perfused heart, likely by interfering with voltage sensing. Thus, SQOR is proposed to contribute to voltage sensing by the mPTP and may be a component of the voltage sensing apparatus that modulates the gating potential of the mPTP.


Subject(s)
Mitochondria, Heart , Mitochondrial Permeability Transition Pore , Oxidoreductases Acting on Sulfur Group Donors , Animals , Mice , Alzheimer Disease , Brain Injuries, Traumatic , Sulfides , Oxidoreductases Acting on Sulfur Group Donors/genetics
2.
Physiol Rep ; 10(15): e15402, 2022 08.
Article in English | MEDLINE | ID: mdl-35923108

ABSTRACT

Infants and children are vulnerable to developing propofol infusion syndrome (PRIS) and young age is a risk factor. Cardiac involvement is often prominent and associated with death. However, the mechanisms of pediatric PRIS are poorly understood because of the paucity of investigation and lack of a gold standard animal model. Unfortunately, in vivo modeling of PRIS in a newborn mouse is not feasible and would be complicated by confounders. Thus, we focused on propofol-induced cardiotoxicity and aimed to develop an ex-vivo model in the isolated-perfused newborn mouse heart. We hypothesized that the model would recapitulate the key cardiac features of PRIS seen in infants and children and would corroborate prior in vitro observations. Isolated perfused newborn mouse hearts were exposed to a toxic dose of propofol or intralipid for 30-min. Surface electrocardiogram, ventricular contractile force, and oxygen extraction were measured over time. Real-time multiphoton laser imaging was utilized to quantify calcein and tetramethylrhodamine ethyl ester fluorescence. Propidium iodide uptake was assessed following drug exposure. A toxic dose of propofol rapidly induced dysrhythmias, depressed ventricular contractile function, impaired the mitochondrial membrane potential, and increased open probability of the permeability transition pore in propofol-exposed hearts without causing cell death. These features mimicked the hallmarks of pediatric PRIS and corroborated prior observations made in isolated newborn cardiomyocyte mitochondria. Thus, acute propofol-induced cardiotoxicity in the isolated-perfused developing mouse heart may serve as a relevant ex-vivo model for pediatric PRIS.


Subject(s)
Propofol , Animals , Animals, Newborn , Arrhythmias, Cardiac , Cardiotoxicity , Heart/physiology , Humans , Mice , Myocytes, Cardiac , Propofol/adverse effects
3.
J Vis Exp ; (184)2022 06 01.
Article in English | MEDLINE | ID: mdl-35723461

ABSTRACT

The mitochondrial permeability transition pore (mPTP) is a voltage-gated, nonselective, inner mitochondrial membrane (IMM) mega-channel important in health and disease. The mPTP mediates leakage of protons across the IMM during low-conductance opening and is specifically inhibited by cyclosporine A (CsA). Coenzyme Q (CoQ) is a regulator of the mPTP, and tissue-specific differences have been found in CoQ content and open probability of the mPTP in forebrain and heart mitochondria in a newborn mouse model of fragile X syndrome (FXS, Fmr1 knockout). We developed a technique to determine the voltage threshold for mPTP opening in this mutant strain, exploiting the role of the mPTP as a proton leak channel. To do so, oxygen consumption and membrane potential (ΔΨ) were simultaneously measured in isolated mitochondria using polarography and a tetraphenylphosphonium (TPP+) ion-selective electrode during leak respiration. The threshold for mPTP opening was determined by the onset of CsA-mediated inhibition of proton leak at specific membrane potentials. Using this approach, differences in voltage gating of the mPTP were precisely defined in the context of CoQ excess. This novel technique will permit future investigation for enhancing the understanding of physiological and pathological regulation of low-conductance opening of the mPTP.


Subject(s)
Mitochondrial Permeability Transition Pore , Ubiquinone , Animals , Mice , Calcium/metabolism , Cyclosporine/pharmacology , Fragile X Mental Retardation Protein , Mitochondria, Heart/metabolism , Mitochondrial Membrane Transport Proteins/physiology , Probability , Protons , Reactive Oxygen Species/metabolism
4.
Mitochondrion ; 65: 1-10, 2022 07.
Article in English | MEDLINE | ID: mdl-35500860

ABSTRACT

Brown adipose tissue (BAT) mitochondria generate heat via uncoupled respiration due to excessive proton leak through uncoupling proteins (UCPs). We previously found hyperthermia in a newborn mouse model of fragile X syndrome and excessive leak in Fmr1 KO forebrain mitochondria caused by CoQ deficiency. The inefficient thermogenic nature of Fmr1 mutant forebrain mitochondria was reminiscent of BAT metabolic features. Thus, we aimed to characterize BAT mitochondrial function in these hyperthermic mice using a top-down approach. Although there was no change in steady-state levels of UCP1 expression between strains, BAT weighed significantly less in Fmr1 mutants compared with controls. Fmr1 KO BAT mitochondria demonstrated impaired substrate oxidation, lower mitochondrial membrane potentials and rates of respiration, and CoQ deficiency. The CoQ analog decylubiquinone normalized CoQ-dependent electron flux and unmasked excessive proton leak. Unlike mutant forebrain, where such deficiency resulted in pathological proton leak, CoQ deficiency within BAT mitochondria resulted largely in abnormal substrate oxidation. This suggests that CoQ is important in BAT for uncoupled respiration to produce heat during development. Although our data provide further evidence of a link between fragile X mental retardation protein (FMRP) and CoQ biosynthesis, the results highlight the importance of CoQ in developing tissues and suggest tissue-specific differences from CoQ deficiency. Because BAT mitochondria are primarily responsible for regulating core body temperature, the defects we describe in Fmr1 KOs could manifest as an adaptive downregulated response to hyperthermia or could result from FMRP deficiency directly.


Subject(s)
Adipose Tissue, Brown , Fragile X Syndrome , Adipose Tissue, Brown/metabolism , Animals , Ataxia , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Mice , Mice, Knockout , Mitochondria/metabolism , Mitochondrial Diseases , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Muscle Weakness , Protons , Ubiquinone/deficiency
5.
Pediatr Res ; 92(5): 1341-1349, 2022 11.
Article in English | MEDLINE | ID: mdl-35173299

ABSTRACT

BACKGROUND: Propofol infusion syndrome (PRIS) is a potentially lethal consequence of long-term propofol administration. Children are vulnerable and cardiac involvement is often prominent and associated with mortality. We aimed to determine the mechanism of propofol toxicity in newborn mice, hypothesizing that propofol would induce discrete defects within immature cardiac mitochondria. METHODS: Newborn murine cardiac mitochondria were exposed to propofol or intralipid in vitro. Non-exposed mitochondria served as controls. Mitochondrial respiration and membrane potential (ΔΨ) were measured and respiratory chain complex kinetics were determined. RESULTS: Propofol and intralipid exerted biological activity in isolated mitochondria. Although intralipid effects were a potential confounder, we found that propofol induced a dose-dependent increase in proton leak and caused a defect in substrate oxidation at coenzyme Q (CoQ). These impairments prevented propofol-exposed cardiomyocyte mitochondria from generating an adequate ΔΨ. The addition of the quinone analog, CoQ0, blocked propofol-induced leak and increased Complex II+III activity. CONCLUSIONS: Propofol uncoupled immature cardiomyocyte mitochondria by inducing excessive CoQ-sensitive leak and interfered with electron transport at CoQ. The findings provide new insight into the mechanisms of propofol toxicity in the developing heart and may help explain why children are vulnerable to developing PRIS. IMPACT: Propofol uncouples immature cardiomyocyte mitochondria by inducing excessive coenzyme Q (CoQ)-sensitive proton leak. Propofol also interferes with electron transport at the level of CoQ. These defects provide new insight into propofol toxicity in the developing heart.


Subject(s)
Mitochondria, Heart , Propofol , Mice , Animals , Mitochondria, Heart/metabolism , Ubiquinone/pharmacology , Ubiquinone/metabolism , Propofol/toxicity , Protons , Oxidation-Reduction
6.
J Neurosurg Anesthesiol ; 34(1): 113-115, 2022 Jan 01.
Article in English | MEDLINE | ID: mdl-34870632

ABSTRACT

Translational science seeks to accelerate the multi-step process by which scientific discoveries are transformed into therapies that can improve the health of individuals and their communities. To facilitate crossing the traditional boundaries between basic and clinical research for instance, a systematic understanding of the scientific and operational principles that underlie each step of the translational cycle is developed to identify and address barriers to translation. Skills required by translational scientists, such as being systems thinkers and process innovators, overlap with those of anesthesiologists, and therefore, it is no surprise that anesthesiologists have contributed to this field. Indeed, the safety and efficacy of anesthesia care has greatly evolved over many decades because anesthesiologists have recognized the importance of readily incorporating physiological and pharmacological basic research findings into clinical practice. This article highlights the characteristics that make anesthesiologists well suited to be translational scientists. We also discuss one example of anesthesiology contributing to the field of translational science during the COVID-19 pandemic. We show that anesthesiologists, regardless of their specific clinical or research interests, have the skill set to become effective and critical players in the field of translational science and emphasize the importance of continued leadership in this field to academic anesthesiology.


Subject(s)
Anesthesiology , COVID-19 , Anesthesiologists , Humans , Pandemics , SARS-CoV-2 , Translational Science, Biomedical
8.
Pediatr Res ; 89(3): 456-463, 2021 02.
Article in English | MEDLINE | ID: mdl-32674111

ABSTRACT

BACKGROUND: Mitochondrial permeability transition pore (mPTP) closure triggers cardiomyocyte differentiation during development while pathological opening causes cell death during myocardial ischemia-reperfusion and heart failure. Ubiquinone modulates the mPTP; however, little is known about its mechanistic role in health and disease. We previously found excessive proton leak in newborn Fmr1 KO mouse forebrain caused by ubiquinone deficiency and increased open mPTP probability. Because of the physiological differences between the heart and brain during maturation, we hypothesized that developing Fmr1 KO cardiomyocyte mitochondria would demonstrate dissimilar features. METHODS: Newborn male Fmr1 KO mice and controls were assessed. Respiratory chain enzyme activity, ubiquinone content, proton leak, and oxygen consumption were measured in cardiomyocyte mitochondria. Cardiac function was evaluated via echocardiography. RESULTS: In contrast to controls, Fmr1 KO cardiomyocyte mitochondria demonstrated increased ubiquinone content and decreased proton leak. Leak was cyclosporine (CsA)-sensitive in controls and CsA-insensitive in Fmr1 KOs. There was no difference in absolute mitochondrial respiration or cardiac function between strains. CONCLUSION: These findings establish the newborn Fmr1 KO mouse as a novel model of excess ubiquinone and closed mPTP in the developing heart. Such a model may help provide insight into the biology of cardiac development and pathophysiology of neonatal heart failure. IMPACT: Ubiquinone is in excess and the mPTP is closed in the developing FXS heart. Strengthens evidence of open mPTP probability in the normally developing postnatal murine heart and provides new evidence for premature closure of the mPTP in Fmr1 mutants. Establishes a novel model of excess CoQ and a closed pore in the developing heart. Such a model will be a valuable tool used to better understand the role of ubiquinone and the mPTP in the neonatal heart in health and disease.


Subject(s)
Disease Models, Animal , Fetal Heart/metabolism , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/metabolism , Mice, Knockout , Mitochondria, Heart/metabolism , Mitochondrial Permeability Transition Pore , Ubiquinone/metabolism , Animals , Atractyloside/analogs & derivatives , Atractyloside/pharmacology , Cyclosporine/pharmacology , Electron Transport , Fragile X Syndrome/genetics , Guanosine Diphosphate/pharmacology , Male , Mice , Mitochondria, Heart/drug effects , Myocytes, Cardiac/metabolism , Oxygen Consumption , Proton-Motive Force , Single-Blind Method , Ubiquinone/analogs & derivatives , Ubiquinone/pharmacology
9.
FASEB J ; 34(6): 7404-7426, 2020 06.
Article in English | MEDLINE | ID: mdl-32307754

ABSTRACT

Fragile X syndrome (FXS) is the leading known inherited intellectual disability and the most common genetic cause of autism. The full mutation results in transcriptional silencing of the Fmr1 gene and loss of fragile X mental retardation protein (FMRP) expression. Defects in neuroenergetic capacity are known to cause a variety of neurodevelopmental disorders. Thus, we explored the integrity of forebrain mitochondria in Fmr1 knockout mice during the peak of synaptogenesis. We found inefficient thermogenic respiration due to futile proton leak in Fmr1 KO mitochondria caused by coenzyme Q (CoQ) deficiency and an open cyclosporine-sensitive channel. Repletion of mitochondrial CoQ within the Fmr1 KO forebrain closed the channel, blocked the pathological proton leak, restored rates of protein synthesis during synaptogenesis, and normalized the key phenotypic features later in life. The findings demonstrate that FMRP deficiency results in inefficient oxidative phosphorylation during the neurodevelopment and suggest that dysfunctional mitochondria may contribute to the FXS phenotype.


Subject(s)
Cell Respiration/physiology , Fragile X Syndrome/metabolism , Fragile X Syndrome/pathology , Mitochondria/metabolism , Mitochondria/pathology , Thermogenesis/physiology , Animals , Autistic Disorder/metabolism , Autistic Disorder/pathology , Disease Models, Animal , Female , Fragile X Mental Retardation Protein/metabolism , Intellectual Disability/metabolism , Intellectual Disability/pathology , Male , Mice , Mice, Knockout , Neurogenesis/physiology , Protons
10.
J Neurosurg Anesthesiol ; 31(1): 163-165, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30767942

ABSTRACT

The potential for long-term neurotoxic effects of anesthetics on the developing human brain has led to intensified research in this area. To date, the human evidence has been inconclusive, but a large body of animal evidence continues to demonstrate cause for concern. On April 14 and 15, 2018 the sixth biennial Pediatric Anesthesia and Neurodevelopmental Assessment (PANDA) study symposium was held at Morgan Stanley Children's Hospital of New York. This symposium brought together clinicians and researchers and served as a platform to review preclinical and clinical data related to anesthesia and neurotoxicity in developing brains. The program participants included many active investigators in the field of anesthesia neurotoxicity as well as stakeholders from different backgrounds with the common interest of potential anesthetic neurotoxicity in children. The moderated poster session included presentations of preclinical animal research studies. These studies focused on defining the anesthetic-induced neurotoxicity phenotype, understanding the mechanism of injury and discovering potential inhibitors of neurotoxic effects.


Subject(s)
Anesthesia/adverse effects , Anesthetics/adverse effects , Developmental Disabilities/chemically induced , Adolescent , Animals , Child , Child, Preschool , Developmental Disabilities/diagnostic imaging , Humans , Infant , Infant, Newborn , Neurotoxicity Syndromes/etiology
11.
Oxid Med Cell Longev ; 2017: 4314025, 2017.
Article in English | MEDLINE | ID: mdl-28630658

ABSTRACT

Autism spectrum disorder (ASD), the fastest growing developmental disability in the United States, represents a group of neurodevelopmental disorders characterized by impaired social interaction and communication as well as restricted and repetitive behavior. The underlying cause of autism is unknown and therapy is currently limited to targeting behavioral abnormalities. Emerging studies suggest a link between mitochondrial dysfunction and ASD. Here, we review the evidence demonstrating this potential connection. We focus specifically on biochemical links, genetic-based associations, non-energy related mechanisms, and novel therapeutic strategies.


Subject(s)
Autism Spectrum Disorder/genetics , Mitochondria/pathology , Autism Spectrum Disorder/pathology , Humans
12.
J Bacteriol ; 195(8): 1735-40, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23396907

ABSTRACT

Germination of Bacillus subtilis spores can be triggered by the binding of specific nutrients, called germinants, to germinant receptors (GRs) in the spore's inner membrane. This interaction eventually initiates, with variable time delays, the release of dipicolinic acid and cations from the spore core--a key step in spore germination. The kinetics of this process are highly heterogeneous for individual spores. In this work, we sought to investigate how the germination heterogeneity was controlled. In particular, we tested whether the rates of germination were determined by GR levels, which vary from spore to spore due to stochastic gene expression. Both the expression levels of GRs and the germination rate were measured in single spores, and the experimental results were compared to theoretical predictions. Our results indicated that the variation in the expression levels of GRs was not the primary factor that controls spore germination heterogeneity. Two alternative hypotheses are discussed in light of this experimental discovery.


Subject(s)
Bacillus subtilis/metabolism , Bacillus subtilis/physiology , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial/physiology , Receptors, Cell Surface/metabolism , Bacillus subtilis/genetics , Bacterial Proteins/genetics , Cell Membrane , Membrane Proteins/genetics , Membrane Proteins/metabolism , Models, Biological , Picolinic Acids , Receptors, Cell Surface/genetics , Spores, Bacterial , Stochastic Processes , Time Factors
13.
J Bacteriol ; 194(21): 5749-58, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22904285

ABSTRACT

As previously reported, gerP Bacillus subtilis spores were defective in nutrient germination triggered via various germinant receptors (GRs), and the defect was eliminated by severe spore coat defects. The gerP spores' GR-dependent germination had a longer lag time between addition of germinants and initiation of rapid release of spores' dipicolinic acid (DPA), but times for release of >90% of DPA from individual spores were identical for wild-type and gerP spores. The gerP spores were also defective in GR-independent germination by DPA with its associated Ca(2+) divalent cation (CaDPA) but germinated better than wild-type spores with the GR-independent germinant dodecylamine. The gerP spores exhibited no increased sensitivity to hypochlorite, suggesting that these spores have no significant coat defect. Overexpression of GRs in gerP spores did lead to faster germination via the overexpressed GR, but this was still slower than germination of comparable gerP(+) spores. Unlike wild-type spores, for which maximal nutrient germinant concentrations were between 500 µM and 2 mM for l-alanine and ≤10 mM for l-valine, rates of gerP spore germination increased up to between 200 mM and 1 M l-alanine and 100 mM l-valine, and at 1 M l-alanine, the rates of germination of wild-type and gerP spores with or without all alanine racemases were almost identical. A high pressure of 150 MPa that triggers spore germination by activating GRs also triggered germination of wild-type and gerP spores identically. All these results support the suggestion that GerP proteins facilitate access of nutrient germinants to their cognate GRs in spores' inner membrane.


Subject(s)
Bacillus subtilis/growth & development , Bacillus subtilis/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Mutation , Spores, Bacterial/growth & development , Spores, Bacterial/genetics , Alanine/metabolism , Calcium/metabolism , Cations, Divalent/metabolism , Hydrostatic Pressure , Picolinic Acids/metabolism , Time Factors , Valine/metabolism
14.
Mol Microbiol ; 81(4): 1061-77, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21696470

ABSTRACT

Dormant bacterial spores are extraordinarily resistant to environmental insults and are vectors of various illnesses. However, spores cannot cause disease unless they germinate and become vegetative cells. The molecular details of initiation of germination are not understood, but proteins essential in early stages of germination, such as nutrient germinant receptors (GRs) and GerD, are located in the spore inner membrane. In this study, we examine how these germination proteins are organized in dormant Bacillus subtilis spores by expressing fluorescent protein fusions that were at least partially functional and observing spores by fluorescence microscopy. We show that GRs and GerD colocalize primarily to a single cluster in dormant spores, reminiscent of the organization of chemoreceptor signalling complexes in Escherichia coli. GRs require all their subunits as well as GerD for clustering, and also require diacylglycerol addition to GerD and GRs' C protein subunits. However, different GRs cluster independently of each other, and GerD forms clusters in the absence of all the GRs. We predict that the clusters represent a functional germination unit or 'germinosome' in the spore inner membrane that is necessary for rapid and cooperative response to nutrients, as conditions known to block nutrient germination also disrupt the protein clusters.


Subject(s)
Bacillus subtilis/chemistry , Bacterial Proteins/analysis , Cell Membrane/chemistry , Spores, Bacterial/chemistry , Genes, Reporter , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Microscopy, Fluorescence , Protein Multimerization , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/genetics
15.
J Biomol Struct Dyn ; 23(6): 667-76, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16615812

ABSTRACT

Divalent metal ions are essential for maintaining functional states of the DNA molecule. Their participation in DNA structure is modulated by the base sequence and varies depending on the nature of the ion. The present investigation addresses the interaction of Ca2+ ions with a tandem repeat of two CA dinucleotides, (CA)2/(TG)2. The binding of Ca2+ to the repeat is monitored by nuclear magnetic resonance (NMR) spectroscopy using chemical shift mapping. Parallel experiments monitor binding of Mg2+ ions to the repeat as well as binding of each ion to a DNA duplex in which the (CA)2/(TG)2 repeat is eliminated. The results reveal that the direction and the magnitude of chemical shift changes induced by Ca2+ ions in the NMR spectra of the repeat are different from those induced by Mg2+ ions. The differences between the two cations are significantly diminished by the elimination of the (CA)2/(TG)2 repeat. These findings suggest a specific interaction of Ca2+ ions with the (CA)2/(TG)2 motif. The specificity of the interaction resides in the two A-T base pairs of the repeat, and it involves the major groove of the first A-T base pair and both grooves of the second A-T base pair.


Subject(s)
Calcium/chemistry , DNA/chemistry , Magnesium/chemistry , Tandem Repeat Sequences , Calcium/metabolism , DNA/metabolism , Magnesium/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...