Top Banner
Cell Metabolism and Cell Death in Neurodegenerative Disease Neurology Clerkship Dr. Ranney Mize Cell Biology and Anatomy Email: [email protected]
30

Cell Metabolism and Cell Death in Neurodegenerative Disease · 2011. 1. 25. · Cell death is a normal process during brain development. It is mostly abnormal in the adult brain.

Feb 04, 2021

Download

Documents

dariahiddleston
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
  • Cell Metabolism and Cell Deathin Neurodegenerative Disease

    Neurology Clerkship

    Dr. Ranney MizeCell Biology and AnatomyEmail: [email protected]

  • • Subcellular organelles in neurons• Protein Synthesis and Degradation• Mitochondria and Oxidative Metabolism• Cell Death: Necrosis and Apoptosis• Pathways leading to cell death• Parkinson’s: Mitochondrial dysfunction+• Alzheimer’s: Protein aggregation+

    Cell Biology and Death of Neurons

  • • Alzheimer’s Disease: abnormal protein cleavage producing toxic protein aggregation + dysfunction of mitochondria

    • Parkinson’s Disease: gene mutations leading to mitochondrial dysfunction + protein aggregation

    • ALS: mutation in superoxide dismutase (SOD1) gene leading to mitochondrial dysfunction, oxidative stress, and protein aggregation

    • Huntington’s Disease: huntingtin gene mutation > polyglutamine repeats > mitochondrial dysfunction, oxidative stress, protein aggregation

    • Stroke, Epilepsy: glutamate excitotoxicity yielding mitochondrial dysfunction + oxidative stress, apoptosis.

    Neurodegenerative Disorders

    PresenterPresentation NotesFocus of this lecture will be on protein aggregation, mitochondrial dysfunction, and oxidative stress.

  • Intracellular Organelles in the Neuron

    Dendrites

    Cell Body

    Dendrites

    Axon

    Synapse

    PresenterPresentation NotesNeuronal morphology varies enormously, with different shapes, sizes (5-50 um), axon projections. Inhibitory vs. excitatory. Dendritic trees are the receptive zones; axons transmit signal to other cells. Nonetheless, soma contents are similar to other cells, including nucleus (gnee transcription), rough ER (protein synthesis), Golgi (transport, routing of proteins), mitochondria (energy source). Other organelles are polyribosomes and smooth ER. Rought ER, mitochondria, are also found in dendrites.

  • 1. Rough Endoplasmic Reticulum (rER): stacks of membrane dotted with ribosomes. Function: synthesis of integral membrane proteins. mRNA transcripts bind to ribosomes which translate the mRNA blueprint in order to assemble proteins from amino acid sequences.2. Mitochondria: membrane bound “kidney” shaped organelles. Function: cellular respiration where converted proteins, fats, sugars (glucose) and oxygen enter the Krebs cycle to produce reactions that yield adenosine triphosphate (ATP). ATP is released into the cytosol as the energy source of the cell.3. Smooth Endoplasmic Reticulum (sER): membrane without ribosomes. Function: varied, including regulation of intracellular concentrations of Ca++.

    Subcellular Organelles of the Neuron

    PresenterPresentation NotesSeveral of the neuron’s organelles are players in the cascade of cell death that occurs after brain injury and disease. Rough ER is a site where proteins are mis-expressed and also a site for failed intracellular Ca++ storage.

  • rER: Protein Synthesis(integral membrane proteins)

    DNA Transcription

    Protein Degradation(ubiquitin-proteosome sys)

    Amino acids

    Proteolysis occurs to break peptide bonds

    via proteases

    requiresenergy

    rER

    Disruption =Protein

    Aggregation

    translocatedthru pore

    Regulatory proteinthat targets proteins to

    the proteosome

    Protein complexwith a core

    (Bear et al, 2007)

    ActivationConjugationRecognition (ligase)

    chaperone

    PresenterPresentation NotesInitial protein synthesis from mRNA code occurs in free polysomes. N terminus then allows signal recognition particle (SRP) to bind to its receptor located on the ER membrane. Protein is then translocated thru an aquaquious pore (TRAM).Ubiquitination refers to the post-translational modification of a protein by the covalent attachment (via an isopeptide bond) of one or more ubiquitin monomers. The most prominent function of ubiquitin is labeling proteins for proteasomal degradation. The main function of the proteasome is to degrade unneeded or damaged proteins by proteolysis, a chemical reaction that breaks peptide bonds. Enzymes that carry out such reactions are called proteases. Proteasomes are part of a major mechanism by which cells regulate the concentration of particular proteins and degrade misfolded proteins, first to peptides, then to constituent amino acids,

  • Mitochondrial Oxidative Metabolism

    I II III IV Velectron chain protein complexes

    NADH

    Cyt c

    (electron transport chain)

    Glucose

    respiration-energy

    ATP

    mtDNA

    complex I

    complex III

    NADHreducing agent > antioxidants. Free radicals incrif blocked.

    Cyt c = heme protein >apoptosis if released

    ( O-2, NO) ( ATP)lipid

    peroxidation

    PresenterPresentation NotesThe pathway for production of ATP is mitochondrial respiration. After glycolysis, the pyruvate product is taken into the mitochondia, oxidatively decarboxylated to acetyl coenzyme-A and further oxidized in the TCA (tricarboxylic acid) cycle. This cycle deposits energy in reduced coenzymes which transfer that energy through what is called the electron transport chain. Energy given to the electrons of the reduced coenzyme NADH by the TCA cycle is transferred in small steps in the inner membrane of the mitochondrion through a chain of five protein complexes. These small oxidation steps accomplish the conversion of ADP to the energy currency molecule ATP. The protein complex ATP synthase makes use of a mitochondrial membrane potential to accomplish the phosphorylation of ADP to ATP Acetyl coenzyme A is an important molecule in metabolism, used to convey carbon atoms within the acetyl group to the citric acid cycle to be oxidized for energy production. NADH = nicotinamide adenine dinucleotide hydrogenase = antioxidant.In metabolism, NAD+ is involved in redox reactions, carrying electrons from one reaction to another. The coenzyme is, therefore, found in two forms in cells: NAD+ is an oxidizing agent – it accepts electrons from other molecules and becomes reduced. This reaction forms NADH, which can then be used as a reducing agent to donate electrons. Complex I = (NADH-ubiquinone reductase), complex III (cytochrome oxidase) and mitochondrial nitric oxide synthase (mtNOS).Mitochondrial complex I (NADH-UQ reductase) catalyzes electron transfer from NADH to ubiquinone and is the molecular pathway to connect the tricarboxylic acid cycle and NADH with the mitochondrial respiratory chain. Complex I is a supra-molecular protein complex of 850 kDa composed of about 40 polypeptide units. peroxynitrite (ONOO-).

  • Cell death is a normal process during brain development.It is mostly abnormal in the adult brain. Neurons die during acute trauma and chronic disease as well as in aging.There are two basic types of cell death: necrotic cell death and programmed cell death.Necrotic cell death occurs most commonly when the brain is damaged by impact injury, subarachnoid hemorrhage, ischemic stroke, seizures, and other sudden insults.Programmed cell death (apoptosis) occurs more commonly in chronic progressive diseases such as Alzheimer’s and Parkinson’s. However, both necrosis and apoptosis can occur in both acute and chronic brain injury. Understanding the mechanisms underlying necrosis and apoptosis has contributed to treatment protocols for these diseases.

    How Do Cells Die

  • Cells Undergoing Necrosis

    1. Necrosis; from Greek for “death”.2. It is a non-programmed cell death

    occurring due to injury or disease= abnormal, inappropriate.

    3. Cell bodies initially swell up leadingto rupture of the cell membrane.

    4. Nucleolus breaks up and nucleusbecomes multi-nucleolated.

    5. Significant cytoplasmic vacuolizationoccurs with breakdown of organelles.

    6. Cell membrane rupture causes releaseof internal contents of the cytoplasm.

    7. This debris produces an inflammatory auto-immune response in brain.

    8. Necrosis is triggered in part by endogenous release of intracellularCa++ produced by ion failure.

    (From Squire et al, 2008)

    vacuoles

    multi-nucleolatednucleus

    darkened chromatin

    swelling, membrane rupture

    http://www.imbb.forth.gr/worms/worms/nrn1174-f1.jpg�

  • Cells Undergoing Apoptosis

    1. Apoptosis; Gr. for “leaves dropping”.2. It is one form of programmed cell death

    (PCD) termed type 1.= normal, appropriate in development.

    3. Cell bodies “round up”, shrink in size.4. Nuclear chromatin condenses, nuclei

    become pyknotic (shrink).5. DNA fragmentation occurs, mediated by

    proteases.6. Eventually the cytoplasm and nucleus

    break up, blebs form on membrane andbud off to form apoptotic bodies.

    7. Residues are phagocytized by glial cells, macrophages.

    8. Triggered by activation of cell-suicidecysteine proteases called caspases.

    (From Squire et al, 2008)

    blebs

    condensed chromatin

    shrinking cell

    apoptotic body

  • Cell Death Summary

    blebs

    apoptotic bodiescondensed chromatin

    dense nucleusApoptosis

    macrophage, lysosomedegradation

    membranerupture

    somatic swellingvacuolization

    phagocytosis

    (From Squire et al, 2008)

    Necrosis

    ‘Normal’ genetic programoccurring in single cells

    Occurs In Multiple Cells > toxic content release which causes brain inflammation

    Toxic releaseof debris

  • • Cell injury (trauma, ischemia, toxins, hypoglycemia) leads to glutamate excitotoxity and significant Ca++ influx into a damaged cell (stroke, status epilepticus)

    • Oxidative stress- ROS generation also lead to release of Ca++ from intracellular stores in the endoplasmic reticulum.

    • DNA damage activates poly ADP-ribose polymerase which decreases pH and leads to acidosis.

    • Increased Ca++ and acidosis produce calpain proteases which together with free radicals rupture the membrane of lysosomes which contain ‘digestive’ enzymes.

    • Release of proteolytic enzymes cathepsin B and D from the lysosome degrade cell structure and metabolism, leading to necrotic cell death.

    Signaling Cascades Leading to Necrosis

    PresenterPresentation NotesPARP is composed of four domains of interest: the DNA binding domain, the cleavage domain, the auto-modification domain, and the catalytic domain. The DNA binding domain is composed of two zinc finger motifs. In the presence of damaged DNA (base pair excised), the DNA binding domain will bind the DNA and induce a conformational shift. It has been shown that this binding occurs independent of the other domains. This is integral in a programmed cell death model based on caspase cleavage inhibition of PARP. The auto-modification domain is responsible for releasing the protein from the DNA after catalysis. It too plays an integral role in cleavage induced inactivation.

  • Pathways to Neuronal Necrosis

    IP3

    ROS overproduction

    Acidosis

    Ca++ Ca++

    Ca++

    Ca++ Ca++

    zinc

    NMDA receptor

    Na+

    (cisteineprotease)

    (aspartylprotease)

    Na2+

    AMPA receptor

    Mg entrypH

    Intracellularcalcium stores

    Poly ADP-ribosepolymerase

    “suicide bag”degrade cell

    organelles

    Glutamate Excitotoxicity(excessive depolarization)

  • • Bcl proteins regulate apoptosis by controlling mitochondrial outer membrane permeabilization, in particular by regulating release of cytochrome c.

    • Anti-apoptotic Bcl-2 proteins in the mitochondrial membrane normally inhibit cytochrome c release.

    • Pro-apoptotic Bcl proteins (Bax, Bid, Bad) translocate to the mitochondrial outer membrane and promote cytochrome c release during injury and disease.

    • Upon release from mitochondria cytochrome c binds to APAF-1 and forms a complex with caspase 9 which is called an ‘apoptosome’.

    • Caspase 9 activates caspase 3 leading to membrane blebbing, chromatin condensation, DNA fragmentation and apoptotic cell death.

    Signaling Cascades In Apoptosis

    PresenterPresentation NotesAPAF-1: Apoptotic protease activating factor 1. Bcl-2 = B cell lymphoma related proteins encoded by Bcl-2 gene.Cytochrome c = protein involved in apoptosis.

  • Intrinsic-Extrinsic Apoptotic Pathways

    (From Bredesen et al, 2006)

    BAX

    BID

    (caspase 3)

    BID = BH3 interacting domain death agonistBAX = Bcl-2–associated pro-apoptotic X- protein

    translocation

    Tissue Necrosis Factor Receptor(TNFR)

    Extrinsic PathwayIntrinsic Pathway

    Mitochondrial MembranePermeabilization

    PresenterPresentation NotesSMAC = second mitochondria-derived activator of caspases.APAF1 = Apoptotic protease activating factorBID = BH3 interacting domain death agonistBAX = Bcl-2–associated pro-apoptotic X- protein

  • • Normal protein degradation involves the ubiquitin-proteosome system for recycling, redistribution of protein. Ubiquitin guides the protein, the proteosome degrades it.

    • The first pathological step blocking protein degradation is the formation of a misfolded protein.

    • Once misfolded, the hydrophobic fragments that are normally buried inside the protein get exposed to the aqueous environment.

    • This intermediate has a high tendency to aggregate and become stabilized because the proteosome is unable to degrade the protein.

    • Incorporation of additional monomers to the protein gives rise to soluable oligomers, protofibrils, and then ‘mature’ protein aggregates which are insoluble.

    Protein Aggregation Pathways

  • oligomers

    fibrils

    Proteolysis occurs to break peptide bonds

    via proteases

    Post-translationalmodification via

    covalent bond

    requiresenergy

    Normal Protein Degradation Abnormal Formation of Aggregates

    aggregates

    Aβ 1-40B sheets

    (cleavage or phosphorylation)

    chaperone

  • • Progressive neurodegenerative disorder affecting 0.5 million Americans, about 1% of the pop. over 60.

    • Symptoms include tremor-at-rest, cogwheel rigidity, postural instability, festinating gate, slow movement(bradykinesia), flat affect, depression.

    • Pathology includes loss of >70% of dopamine neurons in SN compacta and build-up of intracellular inclusions(Lewy bodies) composed of fibrillar α-synuclein.

    • Mitochondrial dysfunction (complex I inhibition leading to oxidative stress) may be the initial step in killing DAcells. However, protein aggregation is also involved.

    • Drug treatments are l-dopa, dopamine agonists, MAOinhibitors. Surgical approach is DBS, usually in the subthalamus.

    Parkinson’s Disease

  • Parkinson’s DiseasePathology

    pill-rolling tremor at rest

    Improved blood flow afterSurgical pallidotomy

    Loss of dopamine inParkinson’s brain

    rigidityakinesia

    Pre Post

    MRI of pathology in

    putamen-globus pallidus

    festinatinggate

    Lewy bodies

    SN

    abnormalposture

  • • Inhibition of complex I of the electron transport chain impairs mitochondrial respiration, including reduction in NADPH and other antioxidants.

    • This leads to increased production of intra- and extra-mitochondrial Reactive Oxygen Species (free radicals such as superoxide anion, hydrogen peroxide, NO).

    • ROS increases the soluble pool of cytochrome c in IMS as well as ROS in the extra-mitochondrial cytosol.

    • Cytosolic ROS produces DNA fragmentation yielding transcriptional upregulation of pro-apoptotic Bax which translocates to the mitochondrial outer membrane.

    • Bax then induces release of cytochrome c which triggers caspase activation and apoptotic cell death.

    Mitochondrial Induced Cell Death Cascade in Parkinson’s

    PresenterPresentation NotesThe molecular mechanisms involved in complex I inactivation include the synergistic inactivations produced by ONOO- mediated reactions, by reactions with free radical intermediates of lipid peroxidation and by amine-aldehyde adduction reactions. The increased rate of O2- generation promotes lipid peroxidation, protein oxidative damage and peroxynitrite (ONOO−) mediated protein nitration (as 3-nitrotyrosine) and nitrosylation (as –S–NO of thiol groups), in a process that lead neurons to apoptosis

  • Perier C et al. PNAS 2007;104:8161-8166

    ©2007 by National Academy of Sciences

    Mitochondrial Dysfunction in Parkinson’s Cell Death

    DNA fragmentation

    MPTP

    up-regulation

    translocation

    oxidative phosphorylation

    (NADH)

    increasedpermeabilization

    transcription

    (Bcl-2 interactingmediator)

    PTP

    Bax, Bim =Pro-apoptoticproteins

    PresenterPresentation NotesFirst step is blocking complex I and hence downregulating NADH. This produces a build-up of reactive oxygen species (ROS). ROS increases cytochrome c from complex III in the intermembrane space (IMS). This cyt c may be released thru a permeability transition pore. Alternatively, cytoplasmic ROS can produce DNA fragmentation which leads to P53 mediated transcription of the pro-apoptotic Bcl-2 protein Bax. Bim then translocates Bax to the mitochondrion, which also permeabilizes the membrane, releases cyto c, activates the caspase reaction leading to apoptosis.MPTP – methyl phenyl tetra-hydropyridine. Proposed pathogenic scenario induced by complex I deficiency with MPTP in which DA neuronal death results from a self-amplifying cascade of deleterious events that start at the mitochondria with the alteration of oxidative phosphorylation and finish at the mitochondria with the activation of the programmed cell death machinery. In this scenario, p53 transcriptionally up-regulates Bax, whereas JNK promotes Bax mitochondrial translocation through transcriptional activation of the BH3-only protein Bim.MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) is a neurotoxin that causes permanent symptoms of Parkinson's disease. The pesticide rotenone has a similar effect.

  • Protein Aggregation in Parkinson’s Disease

    protofibrils

    ROS

    cytochrome c

    caspases

    Gene MutationsProtein Misfolding

    Abnormal ProteinAggregation

    Reactive OxygenSpecies Production

    Oxidative Stress

    Cell Apoptosis

    ATP

    apoptosis

    disrupts proteinguidance

    Proteinstructure

    X

    gene mutationsE3 ubiquitin ligase

    PresenterPresentation NotesLeucine rich repeat kinase-2 may also contribute to proteosomal dysfunction. It is a current hypothesis in the field that cytosolic oxidative stress (increased GSSG and H2O2 levels) favor the post-translational oxidative modification of α-synuclein and its aggregation.

  • Alzheimer’s Disease• Progressive neurodegenerative disease producing severe

    cognitive decline, memory loss, behavioral disorders.• Familial early age Alzheimer’s is an autosomal-dominant

    genetic abnormality in the coding of one or more proteins. Accounts for ~5% of cases. All others are sporadic.

    • Pathology characterized by senile plaques of amyloid-βpeptide and neurofibrillary tangles consisting of hyperphosphorylated tau protein.

    • Genes thought to be involved include amyloid precursor protein (APP) gene and those encoding presenilins 1-2. Presence of the apoE4 gene allele is also a risk factor.

    • Acetylcholine and glutamate transmitter systems are also effected. FDA approved drugs target ACHe (Aricept, an inhibitor), or the NMDA receptor (Namenda, a antagonist)

    • Ultimate neuronal loss occurs first in hippocampus, basal forebrain, temporal and prefrontal cortex, then broadly.

  • • In familial early-onset AD three gene mutations have been identified: amyloid precursor protein (APP) and genes (PS1-PS2) encoding presenilins 1 and 2.

    • Deposits of amyloid β in extracellular space are produced by cleavage of transmembrane APP by β and γ secretases.Aβ-42 is likely the fragment causing amyloid plaques.

    • The abnormal cleavage of APP is produced by a mutation in the presenilin gene. The resulting plaques disrupt cell function.

    • Plaques also increase ROS which produces cell membrane damage and mitrochondrial oxidative stress leading to apoptosis.

    • Neurofibrillary tangles (NFT) also accumulate inside damaged cells. NFT are paired helical filaments consisting of the hyper-phosphorylated microtubule-associated protein tau. These also damage cell function.

    Protein Aggregation and Oxidative Stress in Alzheimer’s Disease

  • Assembly of Plaques and Tangles

    Multi-protein complexwith active docking,

    ATP binding sites

    N-APP

    Amyloid plaques

    NeurofibrillaryTangles

    Tau

    APP Intracellularcytoplasmic domain

    β cleavageenzyme

    BACE = β-site of APP cleaving enzyme

    Amyloid Plaque

    Hyperphosphorylated Tau

  • Cell SignalingPathways in

    Alzheimer’s Disease

    Aβ Protein Aggregation(senile plaques)

    Hyperphosphorylated Tau (neurofibrillary tangles)

    Oxidative Stress

    Apoptosis

    T1 MRI ofAD brain

    ROS Formation

    Bax, CaspasesOxidative degradation

    by free radicals

    ?

  • • Stroke = A “Brain Attack” defined as an “abrupt onset of a focal neurologic deficit”. 3rd leading cause of death inthe US, no. 1 cause of morbidity.

    • Stroke is a vascular insult produced by either obstruction in a blood vessel (embolus, thrombus) or hemorrhage

    (anuerysm, AVM). • Stroke causes ischemia (blood loss), hypoxia ( oxygen)

    and infarct (cell death) leading to paralysis, impaired speech, loss of vision, etc.

    • Tissue damage is due to glutamate excitotoxicity > ionicfailure, excess intracellular Ca+, oxidative stress, etc.

    • Within the infarct, the core is largely necrotic while thesurrounding penumbra can be reperfused. Withoutreperfusion, apoptosis occurs in the penumbra.

    Ischemic and Hemorrhagic Stroke

  • Core and Penumbra in StrokePET

    Ischemic penumbra(perinfarct zone)

    < 20% blood flowdepleted ATPno energy metabolismnecrosis

  • • Glutamate excitotoxicity: ion channel and reuptake failure produces glu overload > prolonged NMDA and AMPA stimulation. This is one source of Ca+ influx.

    • Calcium Dysregulation: Ca+ is also released from mitochondria and ER stores and by activation of Na+/Ca+ exchanger.

    • Oxidative Stress: overload of ROS oxidants from mitochondria (superoxide, hydrogen peroxide, NADPH oxidase, NO) > release of proteases and caspases leading to apoptosis.

    • Spreading Depolarization: triggered by high levels of glu and K+ disrupts ionic gradients yielding massive depolarization of cells.

    • Inflammation: release of cytokines (TNF, IL1β), prosto-glandins, produce severe inflammation in the affected area.

    Detrimental Events in Stroke

  • Imaging the IschemicPenumbra In Stroke

    Moskowitz et al 2010

    Early

    Late

    DWI

    PWI

    Difference

    A

    B

    C

    D

    H2O

    Blood flow

    Slide Number 1Slide Number 2Slide Number 3Slide Number 4Slide Number 5Slide Number 6Slide Number 7Slide Number 8Slide Number 9Slide Number 10Slide Number 11Slide Number 12Slide Number 13Slide Number 14Slide Number 15Slide Number 16Slide Number 17Slide Number 18Slide Number 19Slide Number 20Slide Number 21Slide Number 22Alzheimer’s DiseaseSlide Number 24Slide Number 25Slide Number 26Slide Number 27Slide Number 28Slide Number 29Slide Number 30