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










Publication year range
1.
Cell Rep ; 43(5): 114190, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38717903

ABSTRACT

Neuronal morphology influences synaptic connectivity and neuronal signal processing. However, it remains unclear how neuronal shape affects steady-state distributions of organelles like mitochondria. In this work, we investigated the link between mitochondrial transport and dendrite branching patterns by combining mathematical modeling with in vivo measurements of dendrite architecture, mitochondrial motility, and mitochondrial localization patterns in Drosophila HS (horizontal system) neurons. In our model, different forms of morphological and transport scaling rules-which set the relative thicknesses of parent and daughter branches at each junction in the dendritic arbor and link mitochondrial motility to branch thickness-predict dramatically different global mitochondrial localization patterns. We show that HS dendrites obey the specific subset of scaling rules that, in our model, lead to realistic mitochondrial distributions. Moreover, we demonstrate that neuronal activity does not affect mitochondrial transport or localization, indicating that steady-state mitochondrial distributions are hard-wired by the architecture of the neuron.


Subject(s)
Dendrites , Mitochondria , Animals , Dendrites/metabolism , Mitochondria/metabolism , Drosophila melanogaster/metabolism , Drosophila/metabolism , Neurons/metabolism
2.
Cells ; 12(9)2023 04 29.
Article in English | MEDLINE | ID: mdl-37174687

ABSTRACT

Mitochondrial bioenergetics and dynamics (alterations in morphology and motility of mitochondria) play critical roles in neuronal reactions to varying energy requirements in health and disease. In Alzheimer's disease (AD), mitochondria undergo excessive fission and become less motile. The mechanisms leading to these alterations are not completely clear. Here, we show that collapsin response mediator protein 2 (CRMP2) is hyperphosphorylated in AD and that is accompanied by a decreased interaction of CRMP2 with Drp1, Miro 2, and Mitofusin 2, which are proteins involved in regulating mitochondrial morphology and motility. CRMP2 was hyperphosphorylated in postmortem brain tissues of AD patients, in brain lysates, and in cultured cortical neurons from the double transgenic APP/PS1 mice, an AD mouse model. CRMP2 hyperphosphorylation and dissociation from its binding partners correlated with increased Drp1 recruitment to mitochondria, augmented mitochondrial fragmentation, and reduced mitochondrial motility. (S)-lacosamide ((S)-LCM), a small molecule that binds to CRMP2, decreased its phosphorylation at Ser 522 and Thr 509/514, and restored CRMP2's interaction with Miro 2, Drp1, and Mitofusin 2. This was paralleled by decreased Drp1 recruitment to mitochondria, diminished mitochondrial fragmentation, and improved motility of the organelles. Additionally, (S)-LCM-protected cultured cortical AD neurons from cell death. Thus, our data suggest that CRMP2, in a phosphorylation-dependent manner, participates in the regulation of mitochondrial morphology and motility, and modulates neuronal survival in AD.


Subject(s)
Alzheimer Disease , Animals , Mice , Alzheimer Disease/metabolism , Brain/metabolism , Mice, Transgenic , Mitochondria/metabolism , Neurons/metabolism , Humans
3.
Cells ; 11(10)2022 05 21.
Article in English | MEDLINE | ID: mdl-35626746

ABSTRACT

Recently, we have shown that the physiological roles of a multifunctional protein fructose 1,6-bisphosphatase 2 (FBP2, also called muscle FBP) depend on the oligomeric state of the protein. Here, we present several lines of evidence that in HL-1 cardiomyocytes, a forced, chemically induced reduction in the FBP2 dimer-tetramer ratio that imitates AMP and NAD+ action and restricts FBP2-mitochondria interaction, results in an increase in Tau phosphorylation, augmentation of FBP2-Tau and FBP2-MAP1B interactions, disturbance of tubulin network, marked reduction in the speed of mitochondrial trafficking and increase in mitophagy. These results not only highlight the significance of oligomerization for the regulation of FBP2 physiological role in the cell, but they also demonstrate a novel, important cellular function of this multitasking protein-a function that might be crucial for processes that take place during physiological and pathological cardiac remodeling, and during the onset of diseases which are rooted in the destabilization of MT and/or mitochondrial network dynamics.


Subject(s)
Mitochondria , Myocytes, Cardiac , Microtubules/metabolism , Mitochondria/metabolism , Mitophagy , Myocytes, Cardiac/metabolism , Tubulin/metabolism
4.
Int J Mol Sci ; 23(3)2022 Jan 28.
Article in English | MEDLINE | ID: mdl-35163469

ABSTRACT

Teriflunomide (TFN) limits relapses in relapsing-remitting multiple sclerosis (RRMS) by reducing lymphocytic proliferation through the inhibition of the mitochondrial enzyme dihydroorotate dehydrogenase (DHODH) and the subsequent modulation of de novo pyrimidine synthesis. Alterations of mitochondrial function as a consequence of oxidative stress have been reported during neuroinflammation. Previously, we showed that TFN prevents alterations of mitochondrial motility caused by oxidative stress in peripheral axons. Here, we aimed to validate TFN effects on mitochondria and neuronal activity in hippocampal brain slices, in which cellular distribution and synaptic circuits are largely preserved. TFN effects on metabolism and neuronal activity were investigated by assessing oxygen partial pressure and local field potential in acute slices. Additionally, we imaged mitochondria in brain slices from the transgenic Thy1-CFP/COX8A)S2Lich/J (mitoCFP) mice using two-photon microscopy. Although TFN could not prevent oxidative stress-related depletion of ATP, it preserved oxygen consumption and neuronal activity in CNS tissue during oxidative stress. Furthermore, TFN prevented mitochondrial shortening and fragmentation of puncta-shaped and network mitochondria during oxidative stress. Regarding motility, TFN accentuated the decrease in mitochondrial displacement and increase in speed observed during oxidative stress. Importantly, these effects were not associated with neuronal viability and did not lead to axonal damage. In conclusion, during conditions of oxidative stress, TFN preserves the functionality of neurons and prevents morphological and motility alterations of mitochondria.


Subject(s)
Crotonates/pharmacology , Hippocampus/physiology , Hydrogen Peroxide/adverse effects , Hydroxybutyrates/pharmacology , Mitochondria/metabolism , Nitriles/pharmacology , Oxidative Stress/drug effects , Toluidines/pharmacology , Animals , Energy Metabolism , Hippocampus/drug effects , Male , Mice , Mice, Transgenic , Neurons/drug effects , Neurons/physiology , Oxygen Consumption
5.
Curr Biol ; 31(14): 3098-3114.e7, 2021 07 26.
Article in English | MEDLINE | ID: mdl-34087103

ABSTRACT

Mitochondria supply adenosine triphosphate (ATP) essential for neuronal survival and regeneration. Brain injury and ischemia trigger acute mitochondrial damage and a local energy crisis, leading to degeneration. Boosting local ATP supply in injured axons is thus critical to meet increased energy demand during nerve repair and regeneration in adult brains, where mitochondria remain largely stationary. Here, we elucidate an intrinsic energetic repair signaling axis that boosts axonal energy supply by reprogramming mitochondrial trafficking and anchoring in response to acute injury-ischemic stress in mature neurons and adult brains. P21-activated kinase 5 (PAK5) is a brain mitochondrial kinase with declined expression in mature neurons. PAK5 synthesis and signaling is spatiotemporally activated within axons in response to ischemic stress and axonal injury. PAK5 signaling remobilizes and replaces damaged mitochondria via the phosphorylation switch that turns off the axonal mitochondrial anchor syntaphilin. Injury-ischemic insults trigger AKT growth signaling that activates PAK5 and boosts local energy supply, thus protecting axon survival and facilitating regeneration in in vitro and in vivo models. Our study reveals an axonal mitochondrial signaling axis that responds to injury and ischemia by remobilizing damaged mitochondria for replacement, thereby maintaining local energy supply to support central nervous system (CNS) survival and regeneration.


Subject(s)
Axons , Ischemia , Neurons , Proto-Oncogene Proteins c-akt , p21-Activated Kinases/metabolism , Adenosine Triphosphate , Animals , Cellular Reprogramming , HEK293 Cells , Humans , Mice, Knockout , Mitochondria , Proto-Oncogene Proteins c-akt/metabolism , Regeneration , Signal Transduction
6.
Development ; 148(4)2021 02 19.
Article in English | MEDLINE | ID: mdl-33608250

ABSTRACT

Mitochondria are signaling hubs responsible for the generation of energy through oxidative phosphorylation, the production of key metabolites that serve the bioenergetic and biosynthetic needs of the cell, calcium (Ca2+) buffering and the initiation/execution of apoptosis. The ability of mitochondria to coordinate this myriad of functions is achieved through the exquisite regulation of fundamental dynamic properties, including remodeling of the mitochondrial network via fission and fusion, motility and mitophagy. In this Review, we summarize the current understanding of the mechanisms by which these dynamic properties of the mitochondria support mitochondrial function, review their impact on human cortical development and highlight areas in need of further research.


Subject(s)
Mitochondria/metabolism , Mitochondrial Dynamics/physiology , Mitophagy/physiology , Animals , Apoptosis , Calcium/metabolism , Cerebrum , DNA, Mitochondrial , Dynamins , Humans , Mitochondria/genetics , Mitochondrial Dynamics/genetics , Neurogenesis , Neuroglia , Phenotype , Signal Transduction
7.
Front Cell Dev Biol ; 8: 256, 2020.
Article in English | MEDLINE | ID: mdl-32373611

ABSTRACT

Valosin-containing protein (VCP), also called p97, is an evolutionarily conserved and ubiquitously expressed ATPase with diverse cellular functions. Dominant mutations in VCP are found in a late-onset multisystem degenerative proteinopathy. The neurological manifestations of the disorder include frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS). In these patients, long motor neuron axons could be particularly susceptible to defects in axonal transport. However, whether VCP has a physiological function in maintaining axonal transport and whether this role is impaired by disease-causing mutations remains elusive. Here, by employing live-imaging methods in Drosophila larval axons and performing genetic interaction experiments, we discover that VCP regulates the axonal transport of mitochondria. Downregulation of VCP enhances the retrograde transport of mitochondria and reduces the density of mitochondria in larval axons. This unidirectional motility phenotype is rescued by removing one copy of the retrograde motor dynein heavy chain (DHC), or elevating Miro which facilitates anterograde mitochondrial movement by interacting with the anterograde motor kinesin heavy chain (KHC). Importantly, Miro upregulation also significantly improves ATP production of VCP mutant larvae. We investigate human VCP pathogenic mutations in our fly system. We find that expressing these mutations affects mitochondrial transport in the same way as knocking down VCP. Our results reveal a new role of VCP in mediating axonal mitochondrial transport, and provide evidence implicating impaired mitochondrial motility in the pathophysiology of VCP-relevant neurodegenerative diseases.

8.
Neurobiol Dis ; 127: 410-418, 2019 07.
Article in English | MEDLINE | ID: mdl-30923004

ABSTRACT

Mitochondria are crucial organelles for neurophysiology and brain mitochondrial defects constitute a characteristic of Alzheimer's disease (AD). Impaired axonal mitochondrial traffic, especially the anterograde axonal mitochondrial transport is a pronouncing mitochondrial defect that underlies synaptic failure in AD-related conditions. However, the detailed molecular mechanisms of such axonal mitochondrial abnormality have not been fully understood. KIF5A is a key isoform of kinesin-1, which is a key molecular machinery in facilitating anterograde axonal mitochondrial transport. In this study, we have determined a downregulation of KIF5A in postmortem AD temporal lobes. Further experiments on amyloid beta (Aß)-treated primary neuron culture and 5 × FAD mice suggest a close association of Aß toxicity and KIF5A loss. Downregulation of KIF5A mimics Aß-induced axonal mitochondrial transport deficits, indicating a potential role of KIF5A deficiency in AD-relevant axonal mitochondrial traffic abnormalities. Importantly, the restoration of KIF5A corrects Aß-induced impairments in axonal mitochondrial transport, especially the anterograde traffic, with little or no impact on retrograde axonal mitochondrial motility. Our findings suggest a novel KIF5A-associated mechanism conferring Aß toxicity to axonal mitochondrial deficits. Furthermore, the results implicate a potential therapeutic avenue by protecting KIF5A function for the treatment of AD.


Subject(s)
Amyloid beta-Peptides/metabolism , Axonal Transport/physiology , Axons/metabolism , Kinesins/metabolism , Mitochondria/metabolism , Neurons/metabolism , Animals , Down-Regulation , Kinesins/genetics , Mice , Mice, Knockout
9.
Cond Med ; 1(2): 64-72, 2018.
Article in English | MEDLINE | ID: mdl-30135960

ABSTRACT

Mechanisms of ischemic preconditioning have been extensively studied in gray matter. However, an ischemic episode affects both the gray matter (GM) and white matter (WM) portions of the brain. Inhibition of mitochondrial fission is one of the mechanisms of preconditioning neuronal cell bodies against ischemia. Although axons are anatomical extensions of neuronal cell bodies, injury mechanisms differ between GM and WM. Indeed, axonal dysfunction is responsible for much of the disability associated with clinical deficits observed after stroke; however, the signaling process underlying preconditioning remains unexplored in axons. Using mouse optic nerve, which is a pure isolated WM tract, we show that mitochondria in myelinated axons undergo rapid and profuse fission during oxygen glucose deprivation (OGD) that is mediated by translocation of cytoplasmic Dynamin Related Protein-1 (Drp-1) to mitochondria. OGD-induced mitochondrial fission correlates with reduced mitochondrial motility and loss of axon function. Mitochondrial fragmentation and loss of motility become permanent during the recovery period. Inhibiting mitochondrial fission by administering mitochondrial division inhibitor-1 (Mdivi-1) during OGD preserves mitochondrial shape and motility and promotes axon function recovery. In contrast, preconditioning WM by applying Mdivi-1 only before OGD fails to conserve mitochondrial shape or motility and fails to benefit axon function. Our findings suggest that inhibition of mitochondrial fission during ischemia promotes axon function recovery, but is not sufficient to precondition WM against ischemia. These results raise caution in that approaches to preconditioning neuronal cell bodies may not successfully translate into functional improvement following ischemia.

10.
J Cell Mol Med ; 22(11): 5439-5449, 2018 11.
Article in English | MEDLINE | ID: mdl-30133157

ABSTRACT

Subcellular distribution of mitochondria in neurons is crucial for meeting the energetic demands, as well as the necessity to buffer Ca2+ within the axon, dendrites and synapses. Mitochondrial impairment is an important feature of Parkinson disease (PD), in which both familial parkinsonism genes DJ-1 and PINK1 have a great impact on mitochondrial function. We used differentiated human dopaminergic neuroblastoma cell lines with stable PINK1 or DJ-1 knockdown to study live motility of mitochondria in neurites. The frequency of anterograde and retrograde mitochondrial motility was decreased in PINK1 knockdown cells and the frequency of total mitochondrial motility events was reduced in both cell lines. However, neither the distribution nor the size of mitochondria in the neurites differed from the control cells even after downregulation of the mitochondrial fission protein, Drp1. Furthermore, mitochondria from PINK1 knockdown cells, in which motility was most impaired, had increased levels of GSK3ßSer9 and higher release of mitochondrial Ca2+ when exposed to CCCP-induced mitochondrial uncoupling. Further analysis of the ER-mitochondria contacts involved in Ca2+ shuttling showed that PINK1 knockdown cells had reduced contacts between the two organelles. Our results give new insight on how PINK1 and DJ-1 influence mitochondria, thus providing clues to novel PD therapies.


Subject(s)
Mitochondria/genetics , Parkinson Disease/genetics , Protein Deglycase DJ-1/genetics , Protein Kinases/genetics , Axons/metabolism , Axons/pathology , Calcium/metabolism , Cell Line , Cell Movement , Dendrites/metabolism , Dendrites/pathology , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Dynamins , GTP Phosphohydrolases/genetics , Gene Knockdown Techniques , Glycogen Synthase Kinase 3 beta/genetics , Humans , Microscopy, Electron , Microtubule-Associated Proteins/genetics , Mitochondria/pathology , Mitochondria/ultrastructure , Mitochondrial Proteins/genetics , Neurites/metabolism , Neurites/ultrastructure , Neuroblastoma/genetics , Neuroblastoma/pathology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Synapses/genetics
11.
Cell Physiol Biochem ; 44(4): 1445-1459, 2017.
Article in English | MEDLINE | ID: mdl-29190615

ABSTRACT

Glycogen synthase kinase-3ß (GSK-3ß), a serine/threonine protein kinase, has been reported to show essential roles in molecular pathophysiology of many diseases. Mitochondrion is a dynamic organelle for producing cellular energy and determining cell fates. Stress-induced translocated GSK-3ß may interact with mitochondrial proteins, including PI3K-Akt, PGC-1α, HK II, PKCε, components of respiratory chain, and subunits of mPTP. Mitochondrial pool of GSK-3ß has been implicated in mediation of mitochondrial functions. GSK-3ß exhibits the regulatory effects on mitochondrial biogenesis, mitochondrial bioenergetics, mitochondrial permeability, mitochondrial motility, and mitochondrial apoptosis. The versatile functions of GSK-3ß might be associated with its wide range of substrates. Accumulative evidence demonstrates that GSK-3ß inactivation may be potentially developed as the promising strategy in management of many diseases, such as Alzheimer's disease (AD) and Parkinson's disease (PD). Intensive efforts have been made for exploring GSK-3ß inhibitors. Natural products provide us a great source for screening new lead compounds in inactivation of GSK-3ß. The key roles of GSK-3ß in mediation of mitochondrial functions are discussed in this review.


Subject(s)
Glycogen Synthase Kinase 3 beta/metabolism , Mitochondria/metabolism , Alzheimer Disease/drug therapy , Animals , Apoptosis/drug effects , Biological Products/pharmacology , Biological Products/therapeutic use , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta/chemistry , Lithium Chloride/pharmacology , Lithium Chloride/therapeutic use , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondrial Proteins/metabolism
12.
Autophagy ; 13(11): 1998-1999, 2017.
Article in English | MEDLINE | ID: mdl-28598236

ABSTRACT

The knowledge gap separating the molecular and cellular underpinnings of Parkinson disease (PD) and its pathology hinders treatment innovation. Adding to this difficulty is the lack of a reliable biomarker for PD. Our previous studies identify a link of 2 PD proteins, PINK1/PRKN Parkin to a mitochondrial motor adaptor RHOT1/Miro-1, which mediates mitochondrial motility and mitophagy. Here we review our recent paper showing that a third PD protein, LRRK2, also targets RHOT1 and regulates mitophagy, and pathogenic LRRK2 disrupts this function. Notably, we discover impairments in RHOT1 and mitophagy in sporadic PD patients with no known genetic backgrounds, pointing to RHOT1-mediated mitophagy as a convergent pathway in PD. This novelty opens new doors in PD research toward RHOT1-based therapy and biomarker development.


Subject(s)
Autophagy , Parkinson Disease/genetics , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Mitochondria , Mitophagy/genetics , Protein Kinases/genetics , Ubiquitin-Protein Ligases/genetics
13.
J Intensive Care Med ; 32(1): 86-94, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27443317

ABSTRACT

Metabolic biomarkers have potentially wider use in disease diagnosis and prognosis as well as in monitoring disease response to treatment. While biomarkers such as interleukins, microRNA, and lactate have been proposed for disease surveillance, there are still conflicting results regarding their clinical utility. Treatment of commonly encountered disease of acute care such as sepsis, trauma, and poisoning often relies on clinical diagnosis and therapy guided by use of surrogate markers of illness severity. The measurement of mitochondrial function, including respiration and motility, may offer superior alternatives to such markers. Assessing mitochondrial function in a clinical context has the potential to impact the area of acute care in terms of diagnosis, prognosis, and treatment. The study of mitochondrial bioenergetics has become critical in understanding the pathophysiology and treatment of complex diseases such as diabetes and cardiovascular disorders.


Subject(s)
Cell Respiration , Critical Care , Critical Illness/therapy , Mitochondria/metabolism , Poisoning/metabolism , Sepsis/metabolism , Wounds and Injuries/metabolism , Biomarkers/metabolism , Humans , Monitoring, Physiologic , Poisoning/physiopathology , Poisoning/therapy , Reactive Oxygen Species/metabolism , Sepsis/physiopathology , Sepsis/therapy , Wounds and Injuries/physiopathology , Wounds and Injuries/therapy
14.
Arch Pharm Res ; 39(8): 1144-50, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27464601

ABSTRACT

Superoxide dismutase 1 (SOD1) is a well-known antioxidant enzyme. Mutation of SOD1 is closely associated with the pathogenesis of neurodegenerative disorders, such as amyotrophic lateral sclerosis and Alzheimer's disease. However, the pathologic pathways linking neurodegenerative diseases with mutation of SOD1 remain elusive. Here, we investigated the motility of SOD1-WT and -G93A (a pathogenic mutant of SOD1), and observed correlation of axonal transport of the mutant protein with mitochondria in primary cultured hippocampal neurons. The SOD1-G93A mutant showed significant accumulation at vGlut1-positive synaptic boutons and in cell bodies, compared to SOD1-WT. The proportions of motile WT and G93A proteins were similar (~30 %) while the motility velocity of SOD1-G93A was significantly slower (~40 %) than that of the WT counterpart. This motility defect of SOD1-G93A was highly correlated with mitochondrial movement. Our results collectively suggest that the SOD1-G93A mutant has a defect in motility that is linked to mitochondrial transport in axons.


Subject(s)
Axons/metabolism , Hippocampus/metabolism , Mitochondria/metabolism , Neurons/metabolism , Superoxide Dismutase/metabolism , Animals , Animals, Newborn , Cells, Cultured , Rats , Rats, Sprague-Dawley
15.
Biotechnol Bioeng ; 112(7): 1395-405, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25678368

ABSTRACT

Current methodologies used for mitochondrial motility analysis tend to either overlook individual mitochondrial tracks or analyze only peripheral mitochondria instead of mitochondria in all regions of the cell. Furthermore, motility analysis of an individual mitochondrion is usually quantified by establishing an arbitrary threshold for "directed" motion. In this work, we created a custom, publicly available computational algorithm based on a previously published approach (Giedt et al., 2012. Ann Biomed Eng 40:1903-1916) in order to characterize the distribution of mitochondrial movements at the whole-cell level, while still preserving information about single mitochondria. Our technique is easy to use, robust, and computationally inexpensive. Images are first pre-processed for increased resolution, and then individual mitochondria are tracked based on object connectivity in space and time. When our method is applied to microscopy fields encompassing entire cells, we reveal that the mitochondrial net distances in fibroblasts follow a lognormal distribution within a given cell or group of cells. The ability to model whole-cell mitochondrial motility as a lognormal distribution provides a new quantitative paradigm for comparing mitochondrial motility in naïve and treated cells. We further demonstrate that microtubule and microfilament depolymerization shift the lognormal distribution in directions which indicate decreased and increased mitochondrial movement, respectively. These findings advance earlier work on neuronal axons (Morris and Hollenbeck, 1993. J Cell Sci 104:917-927) by relating them to a different cell type, applying them on a global scale, and automating measurement of mitochondrial motility in general.


Subject(s)
Actin Cytoskeleton/metabolism , Cell Physiological Phenomena , Computational Biology/methods , Cytological Techniques/methods , Mitochondria/physiology , Movement , Algorithms , Cells, Cultured , Fibroblasts/physiology , Humans
16.
Front Cell Neurosci ; 8: 330, 2014.
Article in English | MEDLINE | ID: mdl-25389385

ABSTRACT

Mitochondrial rho GTPase (Miro) is a mitochondrial outer membrane protein containing two GTPase domains and two helix-loop-helix Ca(2+)-binding domains called EF hands. Pioneering genetic studies in Drosophila first revealed a key function of Miro in regulating the axonal transport of mitochondria, during which Miro forms a multi-protein transport complex with Milton and Kinesin heavy chain (KHC) to link trafficking mitochondria with the microtubule (MT) cytoskeleton. Recent studies showed that through binding to the EF hands of Miro and causing conformational changes of Miro and alteration of protein-protein interactions within the transport complex, Ca(2+) can alter the engagement of mitochondria with the MT/kinesin network, offering one mechanism to match mitochondrial distribution with neuronal activity. Despite the importance of the Miro/Milton/Kinesin complex in regulating mitochondrial transport in metazoans, not all components of the transport complex are conserved in lower organisms, and transport-independent functions of Miro are emerging. Here we review the diverse functions of the evolutionarily conserved Miro proteins that are relevant to the development, maintenance, and functioning of the nervous system and discuss the potential contribution of Miro dysfunction to the pathogenesis of diseases of the nervous system.

17.
Annu Rev Cell Dev Biol ; 30: 357-91, 2014.
Article in English | MEDLINE | ID: mdl-25288115

ABSTRACT

Mitochondria are ancient organelles evolved from bacteria. Over the course of evolution, the behavior of mitochondria inside eukaryotic cells has changed dramatically, and the corresponding machineries that control it are in most cases new inventions. The evolution of mitochondrial behavior reflects the necessity to create a dynamic compartment to integrate the myriad mitochondrial functions with the status of other endomembrane compartments, such as the endoplasmic reticulum, and with signaling pathways that monitor cellular homeostasis and respond to stress. Here we review what has been discovered about the molecular machineries that work together to control the collective behavior of mitochondria in cells, as well as their physiological roles in healthy and disease states.


Subject(s)
Mitochondria/physiology , Mitochondrial Turnover/physiology , Animals , DNA, Mitochondrial/metabolism , Dynamins/physiology , Endoplasmic Reticulum/physiology , GTP Phosphohydrolases/physiology , Homeostasis , Humans , Lipid Metabolism , Microtubule-Associated Proteins/physiology , Mitochondrial Diseases/physiopathology , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/physiology , Protein Conformation , Signal Transduction/physiology
18.
Korean J Physiol Pharmacol ; 16(1): 71-7, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22416223

ABSTRACT

Mitochondrial dynamics and distribution is critical for their role in bioenergetics and cell survival. We investigated the consequence of altered fission/fusion on mitochondrial function and motility in INS-1E rat clonal ß-cells. Adenoviruses were used to induce doxycycline-dependent expression of wild type (WT-Mfn1) or a dominant negative mitofusin 1 mutant (DN-Mfn1). Mitochondrial morphology and motility were analyzed by monitoring mitochondrially-targeted red fluorescent protein. Adenovirus-driven overexpression of WT-Mfn1 elicited severe aggregation of mitochondria, preventing them from reaching peripheral near plasma membrane areas of the cell. Overexpression of DN-Mfn1 resulted in fragmented mitochondria with widespread cytosolic distribution. WT-Mfn1 overexpression impaired mitochondrial function as glucose- and oligomycin-induced mitochondrial hyperpolarization were markedly reduced. Viability of the INS-1E cells, however, was not affected. Mitochondrial motility was significantly reduced in WT-Mfn1 overexpressing cells. Conversely, fragmented mitochondria in DN-Mfn1 overexpressing cells showed more vigorous movement than mitochondria in control cells. Movement of these mitochondria was also less microtubule-dependent. These results suggest that Mfn1-induced hyperfusion leads to mitochondrial dysfunction and hypomotility, which may explain impaired metabolism-secretion coupling in insulin-releasing cells overexpressing Mfn1.

19.
Article in English | WPRIM (Western Pacific) | ID: wpr-727555

ABSTRACT

Mitochondrial dynamics and distribution is critical for their role in bioenergetics and cell survival. We investigated the consequence of altered fission/fusion on mitochondrial function and motility in INS-1E rat clonal beta-cells. Adenoviruses were used to induce doxycycline-dependent expression of wild type (WT-Mfn1) or a dominant negative mitofusin 1 mutant (DN-Mfn1). Mitochondrial morphology and motility were analyzed by monitoring mitochondrially-targeted red fluorescent protein. Adenovirus-driven overexpression of WT-Mfn1 elicited severe aggregation of mitochondria, preventing them from reaching peripheral near plasma membrane areas of the cell. Overexpression of DN-Mfn1 resulted in fragmented mitochondria with widespread cytosolic distribution. WT-Mfn1 overexpression impaired mitochondrial function as glucose- and oligomycin-induced mitochondrial hyperpolarization were markedly reduced. Viability of the INS-1E cells, however, was not affected. Mitochondrial motility was significantly reduced in WT-Mfn1 overexpressing cells. Conversely, fragmented mitochondria in DN-Mfn1 overexpressing cells showed more vigorous movement than mitochondria in control cells. Movement of these mitochondria was also less microtubule-dependent. These results suggest that Mfn1-induced hyperfusion leads to mitochondrial dysfunction and hypomotility, which may explain impaired metabolism-secretion coupling in insulin-releasing cells overexpressing Mfn1.


Subject(s)
Animals , Rats , Adenoviridae , Cell Membrane , Cell Survival , Cytosol , Energy Metabolism , Insulin , Insulin-Secreting Cells , Luminescent Proteins , Mitochondria , Mitochondrial Dynamics
20.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-432543

ABSTRACT

Objective The purpose of this study was to investigate the expression of mitochondrial motility-related gene mirol and mitochondrial energy metabolism during an acute bout of prolonged exercise. Methods CS7 BL/6 mice underwent a moderate intensity treadmill running with the slope of 0° and at the speed of 13m/min, and they were randomly divided into 5 groups:resting group(R),exercise groups running for 30min(E30),60min(E60),90min (E90) and 120min(E120),respectively. Respiratory control index and ATP synthesis activity in isolated mitochondria were detected. Skeletal muscle H_2O_2 concentration,mirol mRNA expressions were also measured. Results (1)mirol mRNA contents were significantly increased in groups E30 to E120,as compared with the group R;and mirol mRNA expression increased 32.8%, 107.6%,63.8% and 44.8% respectively in groups E30 to E120. (2)H_2O_2 contents of skeletal muscle were clearly increased in group s E30 to E120. (3)ATP synthesis activity elevated at 30 min of exercise and returned to the baseline thereafter,whereas respiratory control index without remarkable change in groups E30 to E120. Conclusion Muscular mirol mRNA expression significantly increased during 120min of exercise as compared with the resting status and thus facilitated the mitochondrial respiratory and ATP synthesis for matching the energy demand during exercise.

SELECTION OF CITATIONS
SEARCH DETAIL
...