Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Cells ; 10(8)2021 08 10.
Article in English | MEDLINE | ID: mdl-34440815

ABSTRACT

In Alzheimer's disease (AD), the molecular mechanisms involved in the neurodegeneration are still incompletely defined, though this aspect is crucial for a better understanding of the malady and for devising effective therapies. Mitochondrial dysfunctions and altered Ca2+ signaling have long been implicated in AD, though it is debated whether these events occur early in the course of the pathology, or whether they develop at late stages of the disease and represent consequences of different alterations. Mitochondria are central to many aspects of cellular metabolism providing energy, lipids, reactive oxygen species, signaling molecules for cellular quality control, and actively shaping intracellular Ca2+ signaling, modulating the intensity and duration of the signal itself. Abnormalities in the ability of mitochondria to take up and subsequently release Ca2+ could lead to changes in the metabolism of the organelle, and of the cell as a whole, that eventually result in cell death. We sought to investigate the role of mitochondria and Ca2+ signaling in a model of Familial Alzheimer's disease and found early alterations in mitochondria physiology under stressful condition, namely, reduced maximal respiration, decreased ability to sustain membrane potential, and a slower return to basal matrix Ca2+ levels after a mild excitotoxic stimulus. Treatment with an inhibitor of the permeability transition pore attenuated some of these mitochondrial disfunctions and may represent a promising tool to ameliorate mitochondria and cellular functioning in AD and prevent or slow down cell loss in the disease.


Subject(s)
Calcium/metabolism , Hippocampus/metabolism , Mitochondria/metabolism , Neurotoxins/pharmacology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Cells, Cultured , Cyclosporine/pharmacology , Disease Models, Animal , Electron Transport/drug effects , Glutamic Acid/pharmacology , Hippocampus/cytology , Hippocampus/drug effects , Ions/chemistry , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Reactive Oxygen Species/metabolism
2.
BMC Biol ; 19(1): 57, 2021 03 24.
Article in English | MEDLINE | ID: mdl-33761951

ABSTRACT

BACKGROUND: Mitochondrial dysfunction is a common feature of aging, neurodegeneration, and metabolic diseases. Hence, mitotherapeutics may be valuable disease modifiers for a large number of conditions. In this study, we have set up a large-scale screening platform for mitochondrial-based modulators with promising therapeutic potential. RESULTS: Using differentiated human neuroblastoma cells, we screened 1200 FDA-approved compounds and identified 61 molecules that significantly increased cellular ATP without any cytotoxic effect. Following dose response curve-dependent selection, we identified the flavonoid luteolin as a primary hit. Further validation in neuronal models indicated that luteolin increased mitochondrial respiration in primary neurons, despite not affecting mitochondrial mass, structure, or mitochondria-derived reactive oxygen species. However, we found that luteolin increased contacts between mitochondria and endoplasmic reticulum (ER), contributing to increased mitochondrial calcium (Ca2+) and Ca2+-dependent pyruvate dehydrogenase activity. This signaling pathway likely contributed to the observed effect of luteolin on enhanced mitochondrial complexes I and II activities. Importantly, we observed that increased mitochondrial functions were dependent on the activity of ER Ca2+-releasing channels inositol 1,4,5-trisphosphate receptors (IP3Rs) both in neurons and in isolated synaptosomes. Additionally, luteolin treatment improved mitochondrial and locomotory activities in primary neurons and Caenorhabditis elegans expressing an expanded polyglutamine tract of the huntingtin protein. CONCLUSION: We provide a new screening platform for drug discovery validated in vitro and ex vivo. In addition, we describe a novel mechanism through which luteolin modulates mitochondrial activity in neuronal models with potential therapeutic validity for treatment of a variety of human diseases.


Subject(s)
Endoplasmic Reticulum/drug effects , Luteolin/pharmacology , Mitochondria/drug effects , Neurons/metabolism , Animals , Cell Line, Tumor , Drug Evaluation, Preclinical , Endoplasmic Reticulum/metabolism , High-Throughput Screening Assays , Humans , Mice , Mitochondria/metabolism , Neurons/drug effects , Signal Transduction
3.
Aging Clin Exp Res ; 33(6): 1705-1708, 2021 Jun.
Article in English | MEDLINE | ID: mdl-31606858

ABSTRACT

Alzheimer's disease (AD) is the most frequent cause of dementia in the elderly. Few cases are familial (FAD), due to autosomal dominant mutations in presenilin-1 (PS1), presenilin-2 (PS2) or amyloid precursor protein (APP). The three proteins are involved in the generation of amyloid-beta (Aß) peptides, providing genetic support to the hypothesis of Aß pathogenicity. However, clinical trials focused on the Aß pathway failed in their attempt to modify disease progression, suggesting the existence of additional pathogenic mechanisms. Ca2+ dysregulation is a feature of cerebral aging, with an increased frequency and anticipated age of onset in several forms of neurodegeneration, including AD. Interestingly, FAD-linked PS1 and PS2 mutants alter multiple key cellular pathways, including Ca2+ signaling. By generating novel tools for measuring Ca2+ in living cells, and combining different approaches, we showed that FAD-linked PS2 mutants significantly alter cell Ca2+ signaling and brain network activity, as summarized below.


Subject(s)
Alzheimer Disease , Aged , Alzheimer Disease/genetics , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Brain/metabolism , Homeostasis , Humans , Presenilin-1/genetics , Presenilin-1/metabolism , Presenilin-2/genetics , Presenilin-2/metabolism
4.
Cells ; 9(10)2020 09 25.
Article in English | MEDLINE | ID: mdl-32992716

ABSTRACT

Presenilin-2 (PS2) is one of the three proteins that are dominantly mutated in familial Alzheimer's disease (FAD). It forms the catalytic core of the γ-secretase complex-a function shared with its homolog presenilin-1 (PS1)-the enzyme ultimately responsible of amyloid-ß (Aß) formation. Besides its enzymatic activity, PS2 is a multifunctional protein, being specifically involved, independently of γ-secretase activity, in the modulation of several cellular processes, such as Ca2+ signalling, mitochondrial function, inter-organelle communication, and autophagy. As for the former, evidence has accumulated that supports the involvement of PS2 at different levels, ranging from organelle Ca2+ handling to Ca2+ entry through plasma membrane channels. Thus FAD-linked PS2 mutations impact on multiple aspects of cell and tissue physiology, including bioenergetics and brain network excitability. In this contribution, we summarize the main findings on PS2, primarily as a modulator of Ca2+ homeostasis, with particular emphasis on the role of its mutations in the pathogenesis of FAD. Identification of cell pathways and molecules that are specifically targeted by PS2 mutants, as well as of common targets shared with PS1 mutants, will be fundamental to disentangle the complexity of memory loss and brain degeneration that occurs in Alzheimer's disease (AD).


Subject(s)
Alzheimer Disease/genetics , Brain/metabolism , Presenilin-1/genetics , Presenilin-2/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/genetics , Brain/pathology , Calcium/metabolism , Calcium Signaling/genetics , Cell Membrane/genetics , Flavin-Adenine Dinucleotide/genetics , Humans , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Mutant Proteins/genetics , Presenilin-2/metabolism
5.
Cell Rep ; 30(7): 2332-2348.e10, 2020 02 18.
Article in English | MEDLINE | ID: mdl-32075767

ABSTRACT

Mitochondria are key organelles for brain health. Mitochondrial alterations have been reported in several neurodegenerative disorders, including Alzheimer's disease (AD), and the comprehension of the underlying mechanisms appears crucial to understand their relationship with the pathology. Using multiple genetic, pharmacological, imaging, and biochemical approaches, we demonstrate that, in different familial AD cell models, mitochondrial ATP synthesis is affected. The defect depends on reduced mitochondrial pyruvate oxidation, due to both lower Ca2+-mediated stimulation of the Krebs cycle and dampened mitochondrial pyruvate uptake. Importantly, this latter event is linked to glycogen-synthase-kinase-3ß (GSK-3ß) hyper-activation, leading, in turn, to impaired recruitment of hexokinase 1 (HK1) to mitochondria, destabilization of mitochondrial-pyruvate-carrier (MPC) complexes, and decreased MPC2 protein levels. Remarkably, pharmacological GSK-3ß inhibition in AD cells rescues MPC2 expression and improves mitochondrial ATP synthesis and respiration. The defective mitochondrial bioenergetics influences glutamate-induced neuronal excitotoxicity, thus representing a possible target for future therapeutic interventions.


Subject(s)
Alzheimer Disease/genetics , Energy Metabolism/genetics , Mitochondria/metabolism , Pyruvic Acid/metabolism , Animals , Disease Models, Animal , Humans , Transfection
6.
Mar Drugs ; 17(8)2019 Aug 17.
Article in English | MEDLINE | ID: mdl-31426471

ABSTRACT

Herein, we report on the synthesis of a small set of linear precursors of an inosine analogue of cyclic ADP-ribose (cADPR), a second messenger involved in Ca2+ mobilization from ryanodine receptor stores firstly isolated from sea urchin eggs extracts. The synthesized compounds were obtained starting from inosine and are characterized by an N1-alkyl chain replacing the "northern" ribose and a phosphate group attached at the end of the N1-alkyl chain and/or 5'-sugar positions. Preliminary Ca2+ mobilization assays, performed on differentiated C2C12 cells, are reported as well.


Subject(s)
Calcium Signaling/drug effects , Calcium/metabolism , Cyclic ADP-Ribose/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Sea Urchins/chemistry , Second Messenger Systems/drug effects , Animals , Cell Differentiation/drug effects , Eggs , Structure-Activity Relationship
7.
Oxid Med Cell Longev ; 2019: 7210892, 2019.
Article in English | MEDLINE | ID: mdl-31316720

ABSTRACT

Metabolic disorders are severe and chronic impairments of the health of many people and represent a challenge for the society as a whole that has to deal with an ever-increasing number of affected individuals. Among common metabolic disorders are Alzheimer's disease, obesity, and type 2 diabetes. These disorders do not have a univocal genetic cause but rather can result from the interaction of multiple genes, lifestyle, and environmental factors. Mitochondrial alterations have emerged as a feature common to all these disorders, underlining perhaps an impaired coordination between cellular needs and mitochondrial responses that could contribute to their development and/or progression.


Subject(s)
Alzheimer Disease/metabolism , Diabetes Mellitus, Type 2/metabolism , Mitochondria/pathology , Obesity/metabolism , Alzheimer Disease/pathology , Animals , Diabetes Mellitus, Type 2/pathology , Humans , Obesity/pathology
8.
Angew Chem Int Ed Engl ; 58(29): 9917-9922, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31132197

ABSTRACT

Ca2+ handling by mitochondria is crucial for cell life and the direct measure of mitochondrial Ca2+ concentration in living cells is of pivotal interest. Genetically-encoded indicators greatly facilitated this task, however they require demanding delivery procedures. On the other hand, existing mitochondria-targeted synthetic Ca2+ indicators are plagued by several drawbacks, for example, non-specific localization, leakage, toxicity. Here we report the synthesis and characterization of a new fluorescent Ca2+ sensor, named mt-fura-2, obtained by coupling two triphenylphosphonium cations to the molecular backbone of the ratiometric Ca2+ indicator fura-2. Mt-fura-2 binds Ca2+ with a dissociation constant of ≈1.5 µm in vitro. When loaded in different cell types as acetoxymethyl ester, the probe shows proper mitochondrial localization and accurately measures matrix [Ca2+ ] variations, proving its superiority over available dyes. We describe the synthesis, characterization and application of mt-fura-2 to cell types where the delivery of genetically-encoded indicators is troublesome.


Subject(s)
Calcium/metabolism , Fluorescent Dyes/therapeutic use , Mitochondria/metabolism , Fluorescent Dyes/metabolism , Humans
9.
Aging Cell ; 18(3): e12924, 2019 06.
Article in English | MEDLINE | ID: mdl-30793475

ABSTRACT

Mitochondrial dysfunction is implicated in most neurodegenerative diseases, including Alzheimer's disease (AD). We here combined experimental and computational approaches to investigate mitochondrial health and bioenergetic function in neurons from a double transgenic animal model of AD (PS2APP/B6.152H). Experiments in primary cortical neurons demonstrated that AD neurons had reduced mitochondrial respiratory capacity. Interestingly, the computational model predicted that this mitochondrial bioenergetic phenotype could not be explained by any defect in the mitochondrial respiratory chain (RC), but could be closely resembled by a simulated impairment in the mitochondrial NADH flux. Further computational analysis predicted that such an impairment would reduce levels of mitochondrial NADH, both in the resting state and following pharmacological manipulation of the RC. To validate these predictions, we utilized fluorescence lifetime imaging microscopy (FLIM) and autofluorescence imaging and confirmed that transgenic AD neurons had reduced mitochondrial NAD(P)H levels at rest, and impaired power of mitochondrial NAD(P)H production. Of note, FLIM measurements also highlighted reduced cytosolic NAD(P)H in these cells, and extracellular acidification experiments showed an impaired glycolytic flux. The impaired glycolytic flux was identified to be responsible for the observed mitochondrial hypometabolism, since bypassing glycolysis with pyruvate restored mitochondrial health. This study highlights the benefits of a systems biology approach when investigating complex, nonintuitive molecular processes such as mitochondrial bioenergetics, and indicates that primary cortical neurons from a transgenic AD model have reduced glycolytic flux, leading to reduced cytosolic and mitochondrial NAD(P)H and reduced mitochondrial respiratory capacity.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Glycolysis , Mitochondria/metabolism , Neurons/metabolism , Neurons/pathology , Systems Biology , Animals , Cells, Cultured , Female , Male , Mice , Microscopy, Fluorescence
10.
J Biol Chem ; 293(44): 17081-17094, 2018 11 02.
Article in English | MEDLINE | ID: mdl-30228190

ABSTRACT

Mitochondrial Ca2+ uptake through the mitochondrial Ca2+ uniporter (MCU) is a tightly controlled process that sustains cell functions mainly by fine-tuning oxidative metabolism to cellular needs. The kinetics of Ca2+ fluxes across the mitochondrial membranes have been studied both in vitro and in vivo for many years, and the discovery of the molecular components of the MCU has further clarified that this Ca2+ uptake mechanism is based on a complex system subject to elaborate layers of controls. Alterations in the speed or capacity of the in-and-out pathways can have detrimental consequences for both the organelle and the cell, impairing cellular metabolism and ultimately causing cell death. Here, we report that pretreatment of deenergized mitochondria with low-micromolar Ca2+ concentrations for a few minutes markedly increases the speed of mitochondrial Ca2+ uptake upon re-addition of an oxidizable substrate. We found that this phenomenon is sensitive to alterations in the level of the MCU modulator proteins mitochondrial calcium uptake 1 (MICU1) and 2 (MICU2), and is accompanied by changes in the association of MICU1-MICU2 complexes with MCU. This increased Ca2+ uptake capacity, occurring under conditions mimicking those during ischemia/reperfusion in vivo, could lead to a massive amount of Ca2+ entering the mitochondrial matrix even at relatively low levels of cytosolic Ca2+ We conclude that the phenomenon uncovered here represents a potential threat of mitochondrial Ca2+ overload to the cell.


Subject(s)
Calcium/metabolism , Cytosol/metabolism , Mitochondria/metabolism , Animals , Biological Transport , Calcium Channels/genetics , Calcium Channels/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , Liver/metabolism , Mice , Mice, Inbred C57BL , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins/metabolism
11.
Adv Exp Med Biol ; 981: 279-322, 2017.
Article in English | MEDLINE | ID: mdl-29594866

ABSTRACT

A typical characteristic of eukaryotic cells compared to prokaryotes is represented by the spatial heterogeneity of the different structural and functional components: for example, most of the genetic material is surrounded by a highly specific membrane structure (the nuclear membrane), continuous with, yet largely different from, the endoplasmic reticulum (ER); oxidative phosphorylation is carried out by organelles enclosed by a double membrane, the mitochondria; in addition, distinct domains, enriched in specific proteins, are present in the plasma membrane (PM) of most cells. Less obvious, but now generally accepted, is the notion that even the concentration of small molecules such as second messengers (Ca2+ and cAMP in particular) can be highly heterogeneous within cells. In the case of most organelles, the differences in the luminal levels of second messengers depend either on the existence on their membrane of proteins that allow the accumulation/release of the second messenger (e.g., in the case of Ca2+, pumps, exchangers or channels), or on the synthesis and degradation of the specific molecule within the lumen (the autonomous intramitochondrial cAMP system). It needs stressing that the existence of a surrounding membrane does not necessarily imply the existence of a gradient between the cytosol and the organelle lumen. For example, the nuclear membrane is highly permeable to both Ca2+ and cAMP (nuclear pores are permeable to solutes up to 50 kDa) and differences in [Ca2+] or [cAMP] between cytoplasm and nucleoplasm are not seen in steady state and only very transiently during cell activation. A similar situation has been observed, as far as Ca2+ is concerned, in peroxisomes.


Subject(s)
Calcium Signaling/physiology , Cyclic AMP/metabolism , Endoplasmic Reticulum/metabolism , Mitochondrial Membranes/metabolism , Nuclear Envelope/metabolism , Animals , Cyclic AMP/genetics , Endoplasmic Reticulum/genetics , Humans , Nuclear Envelope/genetics
12.
Neurotox Res ; 25(1): 13-23, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24155156

ABSTRACT

Dopaminergic neurons of the substantia nigra selectively degenerate over the course of Parkinson's disease. These neurons are also the most heavily pigmented cells of the brain, accumulating the dark pigment neuromelanin over a lifetime. The massive presence of neuromelanin in these brain areas has long been suspected as a key factor involved in the selective vulnerability of neurons. The high concentration of neuromelanin in substantia nigra neurons seems to be linked to the presence of considerable amounts of cytosolic dopamine that have not been sequestered into synaptic vesicles. Over the past few years, studies have uncovered a dual nature of neuromelanin. Intraneuronal neuromelanin can be a protective factor, shielding the cells from toxic effects of redox active metals, toxins, and excess of cytosolic catecholamines. In contrast, neuromelanin released by dying neurons can contribute to the activation of neuroglia triggering the neuroinflammation that characterizes Parkinson's disease. This article reviews recent studies on the molecular aspects of neuromelanin of the human substantia nigra.


Subject(s)
Dopaminergic Neurons/metabolism , Melanins/metabolism , Substantia Nigra/metabolism , Dopaminergic Neurons/chemistry , Humans , Melanins/biosynthesis , Melanins/chemistry , Substantia Nigra/chemistry
13.
J Biol Chem ; 286(48): 41163-41170, 2011 Dec 02.
Article in English | MEDLINE | ID: mdl-21984833

ABSTRACT

We have studied the pathways for Ca(2+) transport in mitochondria of the fruit fly Drosophila melanogaster. We demonstrate the presence of ruthenium red (RR)-sensitive Ca(2+) uptake, of RR-insensitive Ca(2+) release, and of Na(+)-stimulated Ca(2+) release in energized mitochondria, which match well characterized Ca(2+) transport pathways of mammalian mitochondria. Following larger matrix Ca(2+) loading Drosophila mitochondria underwent spontaneous RR-insensitive Ca(2+) release, an event that in mammals is due to opening of the permeability transition pore (PTP). Like the PTP of mammals, Drosophila Ca(2+)-induced Ca(2+) release could be triggered by uncoupler, diamide, and N-ethylmaleimide, indicating the existence of regulatory voltage- and redox-sensitive sites and was inhibited by tetracaine. Unlike PTP-mediated Ca(2+) release in mammals, however, it was (i) insensitive to cyclosporin A, ubiquinone 0, and ADP; (ii) inhibited by P(i), as is the PTP of yeast mitochondria; and (iii) not accompanied by matrix swelling and cytochrome c release even in KCl-based medium. We conclude that Drosophila mitochondria possess a selective Ca(2+) release channel with features intermediate between the PTP of yeast and mammals.


Subject(s)
Calcium/metabolism , Drosophila Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Adenosine Diphosphate/metabolism , Adenosine Diphosphate/pharmacology , Animals , Antifungal Agents/pharmacology , Benzoquinones/pharmacology , Cyclosporine/pharmacology , Drosophila melanogaster , Drug Resistance/drug effects , Drug Resistance/physiology , Indicators and Reagents/pharmacology , Ion Transport/drug effects , Ion Transport/physiology , Mammals/metabolism , Mitochondrial Permeability Transition Pore , Ruthenium Red/pharmacology , Species Specificity , Yeasts/metabolism
14.
Biochim Biophys Acta ; 1797(11): 1775-9, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20633532

ABSTRACT

We have studied mitochondrial Ca(2+) transport and the permeability transition (PT) in the teleost zebrafish (Danio rerio), a key model system for human diseases. Permeabilized zebrafish embryo cells displayed a mitochondrial energy-dependent Ca(2+) uptake system that, like the Ca(2+) uniporter of mammals, was inhibited by ruthenium red. Zebrafish mitochondria underwent a Ca(2+)-dependent PT that displayed Pi-dependent desensitization by cyclosporin A, and responded appropriately to key modulators of the mammalian PT pore (voltage, pH, ubiquinone 0, dithiol oxidants and cross linkers, ligands of the adenine nucleotide translocator, arachidonic acid). Opening of the pore was documented in intact cells, where it led to death that could largely be prevented by cyclosporin A. Our results represent a necessary step toward the use of zebrafish for the screening and validation of PTP inhibitors of potential use in human diseases, as recently shown for collagen VI muscular dystrophy [Telfer et al., 2010].


Subject(s)
Calcium/metabolism , Cell Membrane Permeability/physiology , Embryo, Nonmammalian/metabolism , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Animals , Calcium Channel Blockers/pharmacology , Cell Death/drug effects , Cells, Cultured , Cyclosporine/pharmacology , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/drug effects , Female , Immunosuppressive Agents/pharmacology , Male , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/physiology , Mitochondria/drug effects , Mitochondrial Membrane Transport Proteins/drug effects , Mitochondrial Permeability Transition Pore , Zebrafish
15.
FEBS Lett ; 584(12): 2504-9, 2010 Jun 18.
Article in English | MEDLINE | ID: mdl-20398660

ABSTRACT

Regulated permeability changes have been detected in mitochondria across species. We review here their key features, with the goal of assessing whether a "permeability transition" similar to that observed in higher eukaryotes is present in other species. The recent discoveries (i) that treatment with cyclosporin A (CsA) unmasks an inhibitory site for inorganic phosphate (Pi) [Basso, E., Petronilli, V., Forte, M.A. and Bernardi, P. (2008) Phosphate is essential for inhibition of the mitochondrial permeability transition pore by cyclosporin A and by cyclophilin D ablation. J. Biol. Chem. 283, 26307-26311], the classical inhibitor of the permeability transition of yeast and (ii) that under proper experimental conditions a matrix Ca(2+)-dependence can be demonstrated in yeast as well [Yamada, A., Yamamoto, T., Yoshimura, Y., Gouda, S., Kawashima, S., Yamazaki, N., Yamashita, K., Kataoka, M., Nagata, T., Terada, H., Pfeiffer, D.R. and Shinohara Y. (2009) Ca(2+)-induced permeability transition can be observed even in yeast mitochondria under optimized experimental conditions. Biochim. Biophys. Acta 1787, 1486-1491] suggest that the mitochondrial permeability transition has been conserved during evolution.


Subject(s)
Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Animals , Calcium/metabolism , Cyclophilins/metabolism , Cyclosporine/pharmacology , Humans , Mammals/metabolism , Mitochondria/drug effects , Mitochondrial Permeability Transition Pore , Permeability/drug effects , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/metabolism
16.
Biochim Biophys Acta ; 1797(6-7): 1113-8, 2010.
Article in English | MEDLINE | ID: mdl-20026006

ABSTRACT

Cyclophilins are a family of peptidyl-prolyl cis-trans isomerases whose enzymatic activity can be inhibited by cyclosporin A. Sixteen cyclophilins have been identified in humans, and cyclophilin D is a unique isoform that is imported into the mitochondrial matrix. Here we shall (i) review the best characterized functions of cyclophilin D in mitochondria, i.e. regulation of the permeability transition pore, an inner membrane channel that plays an important role in the execution of cell death; (ii) highlight new regulatory interactions that are emerging in the literature, including the modulation of the mitochondrial F1FO ATP synthase through an interaction with the lateral stalk of the enzyme complex; and (iii) discuss diseases where cyclophilin D plays a pathogenetic role that makes it a suitable target for pharmacologic intervention.


Subject(s)
Cyclophilins/metabolism , Mitochondria/metabolism , Animals , Calcineurin/metabolism , Peptidyl-Prolyl Isomerase F , Cyclophilins/deficiency , Cyclophilins/genetics , Cyclosporine/metabolism , Disease Models, Animal , Humans , Mice , Mice, Knockout , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Mitochondrial Proton-Translocating ATPases/metabolism , Models, Biological , Protein Interaction Domains and Motifs
17.
J Biol Chem ; 284(49): 33982-8, 2009 Dec 04.
Article in English | MEDLINE | ID: mdl-19801635

ABSTRACT

Blue native gel electrophoresis purification and immunoprecipitation of F(0)F(1)-ATP synthase from bovine heart mitochondria revealed that cyclophilin (CyP) D associates to the complex. Treatment of intact mitochondria with the membrane-permeable bifunctional reagent dimethyl 3,3-dithiobis-propionimidate (DTBP) cross-linked CyPD with the lateral stalk of ATP synthase, whereas no interactions with F(1) sector subunits, the ATP synthase natural inhibitor protein IF1, and the ATP/ADP carrier were observed. The ATP synthase-CyPD interactions have functional consequences on enzyme catalysis and are modulated by phosphate (increased CyPD binding and decreased enzyme activity) and cyclosporin (Cs) A (decreased CyPD binding and increased enzyme activity). Treatment of MgATP submitochondrial particles or intact mitochondria with CsA displaced CyPD from membranes and activated both hydrolysis and synthesis of ATP sustained by the enzyme. No effect of CsA was detected in CyPD-null mitochondria, which displayed a higher specific activity of the ATP synthase than wild-type mitochondria. Modulation by CyPD binding appears to be independent of IF1, whose association to ATP synthase was not affected by CsA treatment. These findings demonstrate that CyPD association to the lateral stalk of ATP synthase modulates the activity of the complex.


Subject(s)
Cyclophilins/physiology , Mitochondria, Heart/enzymology , Mitochondria, Liver/enzymology , Mitochondrial Proton-Translocating ATPases/metabolism , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/chemistry , Animals , Binding Sites , Cattle , Cyclohexanes/pharmacology , Peptidyl-Prolyl Isomerase F , Cyclophilins/metabolism , Heart/physiology , Immunoprecipitation , Mice , Mitochondria, Heart/metabolism , Mitochondria, Liver/metabolism , Models, Biological , Protein Binding
18.
Hum Mol Genet ; 18(11): 2024-31, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19293339

ABSTRACT

Ullrich congenital muscular dystrophy (UCMD) and Bethlem myopathy are inherited muscle disorders caused by mutations of genes encoding the extracellular matrix protein collagen VI (ColVI). Mice lacking ColVI (Col6a1(-/-)) display a myopathic phenotype associated with ultrastructural alterations of mitochondria and sarcoplasmic reticulum, mitochondrial dysfunction with abnormal opening of the permeability transition pore (PTP) and increased apoptosis of muscle fibers. Treatment with cyclosporin (Cs) A, a drug that desensitizes the PTP by binding to cyclophilin (Cyp)-D, was shown to rescue myofiber alterations in Col6a1(-/-) mice and in UCMD patients, suggesting a correlation between PTP opening and pathogenesis of ColVI muscular dystrophies. Here, we show that inactivation of the gene encoding for Cyp-D rescues the disease phenotype of ColVI deficiency. In the absence of Cyp-D, Col6a1(-/-) mice show negligible myofiber degeneration, rescue from mitochondrial dysfunction and ultrastructural defects, and normalized incidence of apoptosis. These findings (i) demonstrate that lack of Cyp-D is equivalent to its inhibition with CsA at curing the mouse dystrophic phenotype; (ii) establish a cause-effect relationship between Cyp-D-dependent PTP regulation and pathogenesis of the ColVI muscular dystrophy and (iii) validate Cyp-D and the PTP as pharmacological targets for the therapy of human ColVI myopathies.


Subject(s)
Apoptosis , Collagen Type VI/genetics , Cyclophilins/genetics , Gene Silencing , Mitochondria/enzymology , Muscle Fibers, Skeletal/cytology , Muscular Diseases/physiopathology , Animals , Cells, Cultured , Collagen Type VI/metabolism , Peptidyl-Prolyl Isomerase F , Cyclophilins/metabolism , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , Muscle Fibers, Skeletal/enzymology , Muscle Fibers, Skeletal/metabolism , Muscular Diseases/enzymology , Muscular Diseases/genetics , Muscular Diseases/metabolism
19.
J Neurosci ; 29(8): 2588-96, 2009 Feb 25.
Article in English | MEDLINE | ID: mdl-19244535

ABSTRACT

Cyclophilin D (CypD), a regulator of the mitochondrial membrane permeability transition pore (PTP), enhances Ca(2+)-induced mitochondrial permeabilization and cell death in the brain. However, the role of CypD in hypoxic-ischemic (HI) brain injury at different developmental ages is unknown. At postnatal day (P) 9 or P60, littermates of CypD-deficient [knock-out (KO)], wild-type (WT), and heterozygous mice were subjected to HI, and brain injury was evaluated 7 d after HI. CypD deficiency resulted in a significant reduction of HI brain injury at P60 but worsened injury at P9. After HI, caspase-dependent and -independent cell death pathways were more induced in P9 CypD KO mice than in WT controls, and apoptotic activation was minimal at P60. The PTP had a considerably higher induction threshold and lower sensitivity to cyclosporin A in neonatal versus adult mice. On the contrary, Bax inhibition markedly reduced caspase activation and brain injury in immature mice but was ineffective in the adult brain. Our findings suggest that CypD/PTP is critical for the development of brain injury in the adult, whereas Bax-dependent mechanisms prevail in the immature brain. The role of CypD in HI shifts from a predominantly prosurvival protein in the immature to a cell death mediator in the adult brain.


Subject(s)
Brain Injuries/etiology , Brain Injuries/metabolism , Cyclophilins/physiology , Hypoxia-Ischemia, Brain/complications , Age Factors , Animals , Animals, Newborn , Apoptosis Inducing Factor/metabolism , Brain/metabolism , Brain/pathology , Brain/ultrastructure , Brain Injuries/genetics , Brain Injuries/pathology , Caspases/metabolism , Cell Death/drug effects , Cell Death/physiology , Peptidyl-Prolyl Isomerase F , Cyclophilins/deficiency , Cytochromes c/metabolism , Disease Models, Animal , Disease Progression , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission/methods , Microtubule-Associated Proteins/metabolism , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , Mitochondrial Membranes/physiology , Mitochondrial Membranes/ultrastructure , Peptide Fragments/pharmacology , Proto-Oncogene Proteins/pharmacology , Time Factors , bcl-2-Associated X Protein/metabolism
20.
J Biol Chem ; 283(39): 26307-11, 2008 Sep 26.
Article in English | MEDLINE | ID: mdl-18684715

ABSTRACT

Energized mouse liver mitochondria displayed the same calcium retention capacity (a sensitive measure of the propensity of the permeability transition pore (PTP) to open) irrespective of whether phosphate, arsenate, or vanadate was the permeating anion. Unexpectedly, however, phosphate was specifically required for PTP desensitization by cyclosporin A (CsA) or by genetic inactivation of cyclophilin D (CyP-D). Indeed, when phosphate was replaced by arsenate, vanadate, or bicarbonate, the inhibitory effects of CsA and of CyP-D ablation on the PTP disappeared. After loading with the same amount of Ca(2+) in the presence of arsenate or vanadate but in the absence of phosphate, the sensitivity of the PTP to a variety of inducers was identical in mitochondria from wild-type mice, CyP-D-null mice, and wild-type mice treated with CsA. These findings call for a reassessment of conclusions on the role of the PTP in cell death that are based on the effects of CsA or of CyP-D ablation.


Subject(s)
Cell Membrane Permeability/physiology , Cyclophilins/metabolism , Cyclosporine/pharmacology , Enzyme Inhibitors/pharmacology , Mitochondria, Liver/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/metabolism , Phosphates/metabolism , Animals , Anions/metabolism , Anions/pharmacology , Cell Death/drug effects , Cell Death/physiology , Cell Membrane Permeability/drug effects , Peptidyl-Prolyl Isomerase F , Cyclophilins/genetics , Ion Transport/drug effects , Ion Transport/physiology , Mice , Mice, Knockout , Mitochondria, Liver/genetics , Mitochondrial Proteins/genetics , Phosphates/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL