The diphenylpyrazole compound anle138b blocks Aβ channels and rescues disease phenotypes in a mouse model for amyloid pathology

Ana Martinez Hernandez, Hendrik Urbanke, Alan L Gillman, Joon Lee, Sergey Ryazanov, Hope Y Agbemenyah, Eva Benito, Gaurav Jain, Lalit Kaurani, Gayane Grigorian, Andrei Leonov, Nasrollah Rezaei-Ghaleh, Petra Wilken, Fernando Teran Arce, Jens Wagner, Martin Fuhrmann, Mario Caruana, Angelique Camilleri, Neville Vassallo, Markus Zweckstetter, Roland Benz, Armin Giese, Anja Schneider, Martin Korte, Ratnesh Lal, Christian Griesinger, Gregor Eichele, Andre Fischer, Ana Martinez Hernandez, Hendrik Urbanke, Alan L Gillman, Joon Lee, Sergey Ryazanov, Hope Y Agbemenyah, Eva Benito, Gaurav Jain, Lalit Kaurani, Gayane Grigorian, Andrei Leonov, Nasrollah Rezaei-Ghaleh, Petra Wilken, Fernando Teran Arce, Jens Wagner, Martin Fuhrmann, Mario Caruana, Angelique Camilleri, Neville Vassallo, Markus Zweckstetter, Roland Benz, Armin Giese, Anja Schneider, Martin Korte, Ratnesh Lal, Christian Griesinger, Gregor Eichele, Andre Fischer

Abstract

Alzheimer's disease is a devastating neurodegenerative disease eventually leading to dementia. An effective treatment does not yet exist. Here we show that oral application of the compound anle138b restores hippocampal synaptic and transcriptional plasticity as well as spatial memory in a mouse model for Alzheimer's disease, when given orally before or after the onset of pathology. At the mechanistic level, we provide evidence that anle138b blocks the activity of conducting Aβ pores without changing the membrane embedded Aβ-oligomer structure. In conclusion, our data suggest that anle138b is a novel and promising compound to treat AD-related pathology that should be investigated further.

Keywords: Alzheimer's disease; Aβ channels; amyloid pathology; gene expression; membrane pores.

Conflict of interest statement

AG and CG are co‐founders of MODAG. AL is partly employed by MODAG.

© 2017 The Authors. Published under the terms of the CC BY 4.0 license.

Figures

Figure EV1. Experimental design
Figure EV1. Experimental design
Overview of the experimental approach employed for the pre‐plaque and the post‐plaque group. Red arrows underneath the time line indicate onset of pathology in APPPS1△9 mice.
Figure 1. Anle138b rescues hippocampal LTP deficits…
Figure 1. Anle138b rescues hippocampal LTP deficits spatial memory in the pre‐plaque group
  1. In wild‐type mice of the pre‐plaque group (treated from 2 to 6 months of age with anle138b), robust LTP that lasts for at least 3 h is elicited upon a strong tetanization (STET) (3 trains of 100 pulses at 100 Hz given 10 min apart, arrows) at the Schaffer collateral CA3‐CA1 synapse (t‐test, P = 0.00005; n = 16 comparing before vs. after STET).

  2. LTP is not maintained in APPPS1Δ9 mice treated with placebo. Here, the potentiation declined to baseline after 3 h (t‐test, P = 0.08; n = 20 comparing before vs. after STET).

  3. APPPS1Δ9 treated with anle138b show a rescue of the LTP impairment (t‐test, P = 0.0001; n = 23 comparing before vs. after STET).

  4. Escape latency in the Morris water maze test is impaired in placebo‐ but not in anle138b‐treated APP mice (one‐way ANOVA F = 16.01, **P = 0.0008; n = 15/group).

  5. Probe test performed 24 h after the last training session. The lower panel shows representative swimming path during the probe test. T = target quadrant compared vs. other quadrants (t‐test, ***P = 0.00002; n = 15/group).

  6. Average swim speed during water maze training (n = 15/group).

  7. Explorative behavior in the open field test. Upper panel: Representative motion tracks during the test session. Lower panels show the total distance travelled during the 5‐min test session (n = 15/group).

  8. Bar graph showing the time spent in the center vs. the corner of the open field (n = 15/group).

Data information: Error bars indicate SEM.
Figure 2. Anle138b rescues hippocampal LTP deficits…
Figure 2. Anle138b rescues hippocampal LTP deficits and spatial memory in the post‐plaque group
  1. Wild‐type mice of the post‐plaque group (treated from 6 to 10 months of age) display robust LTP upon STET (arrows) that was maintained throughout the recording session (t‐test, P = 0.00006; n = 30 comparing before vs. after STET).

  2. Lasting LTP induced by STET was not observed in 10‐month‐old APPPS1Δ9 placebo‐treated mice. The potentiation decayed to baseline after 3 h (t‐test, P = 0.16; n = 19 comparing before vs. after STET).

  3. Treatment with anle138b starting at 6 months of age rescues LTP deficit in 10‐month‐old APPPS1Δ9 mice (t‐test, P = 0.0003; n = 20 comparing before vs. after STET).

  4. Escape latency in the Morris water maze test is impaired in placebo‐treated APP mice and partially restored to WT levels in anle138b‐treated APP mice (one‐way ANOVA, F = 35.94, P = 0.00006; *P = 0.0309 for APP + anle138b vs. APP + placebo and P = 0.4 for APP + placebo vs. WT + anle138b; n = 7/group).

  5. Probe test performed 24 h after the last training session. The lower panel shows representative swimming path during the probe test. T = target quadrant compared vs. other quadrants (t‐test, **P = 0.0007, *P = 0.0031; n = 7/group).

  6. Average swim speed during water maze training (n = 7/group).

  7. Explorative behavior in the open field test. Upper panel: Representative motion tracks during the test session. Lower panels show the total distance travelled during the 5‐min test session (n = 7/group).

  8. Bar graph showing the time spent in the center vs. the corner of the open field (n = 7/group).

Data information: Error bars indicate SEM. In panels (A–C), triplets of arrows represent STET applied for inducing L‐LTP. Insets in each graph represent typical fEPSP traces recorded 15 min before (dotted line), 30 min after (broken line), and 3 h after (full line) STET.
Figure 3. Hippocampal transcriptome analysis in anle138b‐treated…
Figure 3. Hippocampal transcriptome analysis in anle138b‐treated mice
  1. Upper panel: Heat map showing differentially expressed genes in placebo‐treated WT (n = 4), placebo‐treated APPPS1ΔE9 mice (n = 4), and anle138b‐treated APPPS1ΔE9 mice (n = 4) of the pre‐plaque group. Note that a WT‐like gene expression profile is to a large extent reinstated in APPPS1ΔE9 mice upon treatment with anle138b. Lower panel: Bar graph showing the number of up and down‐regulated genes.

  2. Pathways down‐regulated (blue) or up‐regulated (red) in 6‐month‐old APPPS1ΔE9 mice when compared to age‐matched WT controls (pre‐plaque group; see Fig EV1).

  3. Expression of APP and PS1 genes in WT and APPPS1ΔE9 transgenic mice, placebo‐ or anle138b‐treated in pre‐ and post‐plaque groups. Note that anle138b treatment does not affect the expression level of APP or PS1. Post hoc analysis revealed a significantly increased expression of APP and PS1 in transgenic mice when compared to wild‐type mice (*P < 0.05). Error bars indicate SEM.

  4. Upper panel: Heat map showing differentially expressed genes in placebo‐treated WT (n = 4), placebo‐treated APPPS1ΔE9 mice (n = 5), and anle138b‐treated APPPS1ΔE9 mice (n = 5) of the post‐plaque group. Lower panel: Bar graph showing the number of up‐ and down‐regulated genes.

  5. Venn diagram showing that 10‐month‐old mice treated with placebo or anle138b show very similar changes in hippocampal gene expression (post‐plaque group; see Fig EV1).

  6. Pathway analysis based on the 103 genes commonly increased in placebo‐ and anle138b‐treated APPPS1ΔE9 mice at 10 months of age. Note that the pathways are exclusively linked to neuroinflammation.

Figure 4. Amyloid plaque pathology is ameliorated…
Figure 4. Amyloid plaque pathology is ameliorated by anle138b treatment in the pre‐plaque (A) and post‐plaque (B) groups
  1. Reduced amyloid pathology as indicated by reduced number of plaques (left panel) and reduced area covered by plaques (right panel) in the pre‐plaque group (t‐test ***P = 0.00003; n = 5/group).

  2. Reduced amyloid pathology as indicated by reduced number of plaques (left panel) and reduced area covered by plaques in the post‐plaque group (t‐test ***P = 0.00007, **P = 0.005; n = 5/group).

  3. Representative images showing thioflavin S staining in the hippocampus and cortex of mice of the pre‐ and post‐plaque groups. Scale bar = 200 μm.

Data information: Error bars indicate SEM.
Figure 5. Anle138b ameliorates Aβ 1‐40 and…
Figure 5. Anle138b ameliorates Aβ1‐40 and Aβ1‐42‐induced membrane integrity
  1. Aβ1‐42 induces pore‐like step ionic current increases across lipid bilayer membranes and grows with inverted sign after voltage inversion to amplifier saturation current. In the presence of anle138b, the current remains at low amplitude and does not increase beyond 30 pA.

  2. Expanded trace of anle138b‐treated membrane presented in (A). Discreet conductance levels are highlighted suggesting multiple opening and closing events for three individual pores.

  3. Hippocampal neurons (DIV 10) were treated with anle138b (1 μM) or vehicle before Aβ1‐40 oligomers or monomers were added (10 μM, n = 4/group). After 48 h, membrane integrity was measured as fluorescence intensity using a CyQUANT assay (Thermo Fisher). In the vehicle group, membrane integrity was significantly impaired when treated with Aβ oligomers comparing with control neurons or Aβ monomer‐treated neurons, anle138b‐treated neurons did not exhibit a difference between addition of Aβ monomers or Aβ oligomers (t‐test, **P = 0.007).

  4. Same experimental setting as in (C) but cell viability was measured using the MTT assay (t‐test, *P = 0.002 vs. control). No difference in cell viability was observed for Aβ monomer or oligomer treatment in the absence or presence of anle138b.

  5. Schematic of potential mechanisms of activity inhibition for anle138b. In the absence of anle138b, Aβ monomers and/or oligomers insert in the membrane and form conducting pores. Treatment with anle138b renders these conductive channels inactive most probably due to reduced life time of the open state.

Data information: Error bars indicate SEM.
Figure EV2. Observation of Aβ pores by…
Figure EV2. Observation of Aβ pores by AFM in anle138b‐doped membranes
AFM tapping mode images showing similar pore structures in membranes with and without anle138b.
  1. A

    POPE/DOPS (1:1) membranes without compound and not exposed to Aβ1‐42 showing lateral phase separation. Scale bar is as indicated and z scale is 5 nm.

  2. B

    Membrane with the same lipid composition as in (A) without compound reconstituted with Aβ1‐42 at a 1:60 peptide/lipid molar ratio. White circles represent the position of pore‐like structures. Scale bar is 100 nm and z scale is 5 nm.

  3. C

    Membrane with the same lipid composition as above with anle138b (10 mM with respect to lipid volume) and reconstituted with Aβ1‐42 (1:60 peptide/lipid molar ratio). White circles represent the position of pore‐like structures. Scale bar is 100 nm and z scale is 5 nm.

  4. D, E

    Four representative pores extracted from (B) and (C), respectively. Image size is 23.4 nm × 23.4 nm.

  5. F

    Table summarizing that Aβ pores show the same dimension in the presence or absence of anle138b. Values are represented as average ±SD.

References

    1. Arce FT, Jang H, Ramachandran S, Landon PB, Nussinov R, Lal R (2011) Polymorphism of amyloid β peptide in different environments: implications for membrane insertion and pore formation. Soft Matter 7: 5267–5273
    1. Arispe N, Rojas E, Pollard HB (1993) Alzheimer disease amyloid beta protein forms calcium channels in bilayer membranes: blockade by tromethamine and aluminum. Proc Natl Acad Sci USA 90: 567–571
    1. Benito E, Urbanke H, Ramachandran B, Barth J, Halder R, Awasthi A, Jain G, Capece V, Burkhardt S, Navarro‐Sala M et al (2015) HDAC inhibitor‐dependent transcriptome and memory reinstatement in cognitive decline models. J Clin Invest 17: 3572–3584
    1. Benz R, Janko K, Boos W, Lauger P (1978) Formation of large, ion‐permeable membrane channels by matrix protein (Porin) of Escherichia‐Coli . Biochem Biophys Acta 511: 305–319
    1. Bieschke J, Russ J, Friedrich RP, Ehrnhoefer DE, Wobst H, Neugebauer K, Wanker EE (2010) EGCG remodels mature alpha‐synuclein and amyloid‐beta fibrils and reduces cellular toxicity. Proc Natl Acad Sci USA 107: 7710–7715
    1. Brand AH, Perrimon N (1993) Targeted gene expression as a means of altering cell fates and generating dominant phenotypes. Development 2: 401‐415
    1. Capone R, Jang H, Kotler SA, Kagan BL, Nussinov R, Lal R (2012) Probing structural features of Alzheimer's amyloid‐beta pores in bilayers using site‐specific amino acid substitutions. Biochemistry 51: 776–785
    1. Connelly L, Arce FT, Jang H, Capone R, Kotler SA, Ramachandran S, Kagan BL, Nussinov R, Lal R (2012a) Atomic force microscopy and MD simulations reveal pore‐like structures of all‐D‐Enantiomer of Alzheimer's β‐amyloid peptide: relevance to the ion channel mechanism of AD pathology. J Phys Chem B 116: 1728–1735
    1. Connelly L, Jang H, Arce FT, Ramachandran S, Kagan BL, Nussinov R, Lal R (2012b) Effects of point substitutions on the structure of toxic Alzheimer's β‐amyloid channels: atomic force microscopy and molecular dynamics simulations. Biochemistry 51: 3031–3038
    1. Crowther DC, Kinghorn KJ, Miranda E, Page R, Curry JA, Duthie FA (2005) Intraneuronal Abeta, non‐amyloid aggregates and neurodegeneration in a Drosophila model of Alzheimer's disease. Neuroscience 132: 123–135
    1. Demuro A, Smith M, Parker I (2011) Single‐channel Ca(2+) imaging implicates Aβ1‐42 amyloid pores in Alzheimer's disease pathology. J Cell Biol 195: 515–524
    1. Diaz JC, Simakova O, Jacobson KA, Arispe N, Pollard HB (2009) Small molecule blockers of the Alzheimer A beta calcium channel potently protect neurons from A beta cytotoxicity. Proc Natl Acad Sci USA 106: 3348–3353
    1. Djebali S, Davis CA, Merkel A, Dobin A, Lassmann T, Mortazavi A, Tanzer A, Lagarde J, Lin WSF, Xue C et al (2012) Landscape of transcription in human cells. Nature 489: 101–108
    1. Doig AJ, Derreumaux P (2015) Inhibition of protein aggregation and amyloid formation by small molecules. Curr Opin Struct Biol 30: 50–56
    1. Ehrnhoefer DE, Bieschke J, Boeddrich A, Herbst M, Masino L, Lurz R, Engemann S, Pastore A, Wanker EE (2008) EGCG redirects amyloidogenic polypeptides into unstructured, off‐pathway oligomers. Nat Struct Mol Biol 15: 558–566
    1. Fischer A (2014a) Epigenetic memory: the Lamarckian brain. EMBO J 33: 945–967
    1. Fischer A (2014b) Targeting histone‐modifications in Alzheimer's disease. What is the evidence that this is a promising therapeutic avenue? Neuropsychopharmacology 80: 95–102
    1. Fischer A (2016) Environmental enrichment as a method to improve cognitive function. What can we learn from animal models? NeuroImage 1: 42–47
    1. Gillman AL, Jang H, Lee J, Ramachandran S, Kagan BL, Nussinov R, Arce FT (2014) Activity and architecture of pyroglutamate modified amyloid‐β (AβpE3‐42) pores. J Phys Chem B 118: 7335–7344
    1. Goate A, Hardy J (2012) Twenty years of Alzheimer's disease‐causing mutations. J Neurochem 120: 3–8
    1. Guntern R, Bouras C, Hof PR, Vallet PG (1992) An improved thioflavine‐s method for staining neurofibrillary tangles and senile plaques in Alzheimers disease. Experientia 48: 8–10
    1. Haass C, Selkoe DJ (2007) Soluble protein oligomers in neurodegeneration: lessons from the Alzheimer's amyloid beta‐peptide. Nat Rev Mol Cell Biol 8: 112–116
    1. Iversen LL, Mortishire‐Smith RJ, Pollack SJ, Shearman MS (1995) The toxicity in vitro of beta‐amyloid protein. Biochem J 311: 1–16
    1. Jakob‐Roetne R, Jacobsen H (2009) Alzheimer's disease: from pathology to therapeutic approaches. Angew Chem Int Ed 48: 3030–3059
    1. Jang H, Zheng J, Lal R, Nussinov R (2008) New structures help the modeling of toxic amyloidβ ion channels. Trends Biochem Sci 33: 91–100
    1. Jankowsky JL, Slunt HH, Ratovitski T, Jenkins NA, Copeland NG, Borchelt DR (2001) Co‐expression of multiple transgenes in mouse CNS: a comparison of strategies. Biomol Eng 17: 157–165
    1. Jankowsky JL, Fadale DJ, Anderson J, Xu GM, Gonzales V, Jenkins NA, Copeland NG, Lee MK, Younkin LH, Wagner SL et al (2004) Mutant presenilins specifically elevate the levels of the 42 residue beta‐amyloid peptide in vivo: evidence for augmentation of a 42‐specific gamma secretase. Hum Mol Genet 13: 159–170
    1. Kummer MP, Hammerschmidt T, Martinez A, Terwel D, Eichele G, Witten A, Figura S, Stoll M, Schwartz S, Pape HC et al (2014) Ear2 deletion causes early memory and learning deficits in APP/PS1 mice. J Neurosci 34: 8845–8854
    1. Lalonde R, Kim HD, Maxwell JA, Fukuchi K (2005) Exploratory activity and spatial learning in 12‐month‐old APP(695)SWE/co+PS1/Delta E9 mice with amyloid plaques. Neurosci Lett 390: 87–92
    1. Lee J, Gillman AL, Jang H, Ramachandran S, Kagan BL, Nussinov R, Arce FT (2014) Role of the fast kinetics of pyroglutamate‐modified amyloid‐β oligomers in membrane binding and membrane permeability. Biochemistry 53: 4704–4714
    1. Lin HAI, Bhatia R, Lal R (2001) Amyloid β protein forms ion channels: implications for Alzheimer's disease pathophysiology. FASEB J 15: 2433–2444
    1. Liu F, Arce FT, Ramachandran S, Lal R (2006) Nanomechanics of hemichannel conformations – Connexin flexibility underlying channel opening and closing. J Biol Chem 281: 23207–23217
    1. Liu M, Chen F, Sha L, Wang S, Tao L, Yao L, He M, Yao Z, Liu H, Zhu Z et al (2014) (‐)‐Epigallocatechin‐3‐gallate ameliorates learning and memory deficits by adjusting the balance of TrkA/p75NTR signaling in APP/PS1 transgenic mice. Mol Neurobiol 49: 1350–1363
    1. Love MI, Huber W, Anders S (2014) Moderated estimation of fold change and dispersion for RNA‐seq data with DESeq2. Genome Biol 15: 550
    1. Matarin M, Salih DA, Yasvoina M, Cummings DM, Guelfi S, Liu W, Nahaboo Solim MA, Moens TG, Paublete RM, Ali SS et al (2015) A genome‐wide gene‐expression analysis and database in transgenic mice during development of amyloid or tau pathology. Cell Rep 10: 633–644
    1. McGuire SE, Le PT, Osborn AJ, Matsumoto K, Davis RL (2003) Spatiotemporal rescue of memory dysfunction in Drosophila . Science 302: 1765–1768
    1. Morris R (1984) Developments of a water‐maze procedure for studying spatial learning in the rat. J Neurosci Methods 11: 47–60
    1. Mueller P, Rudin DO, Tien HT, Wescott WC (1962) Reconstitution of cell membrane structure in vitro and its transformation into an excitable system. Nature 194: 979–980
    1. Querfurth HW, LaFerla FM (2010) Alzheimer's disease. N Engl J Med 362: 329–344
    1. Quist A, Doudevski I, Lin H, Azimova R, Ng D, Frangione B, Kagan B, Ghiso J, Lal R (2005) Amyloid ion channels: a common structural link for protein‐misfolding disease. Proc Natl Acad Sci USA 102: 10427–10432
    1. Reiserer RS, Harrison FE, Syverud DC, McDonald MP (2007) Impaired spatial learning in the APP(Swe)+PSEN1 Delta E9 bigenic mouse model of Alzheimer's disease. Genes Brain Behav 6: 54–65
    1. Sarkar B, Mithu VS, Chandra B, Mandal A, Chandrakesan M, Bhowmik D, Madhu PK, Maiti S (2014) Significant structural differences between transient amyloid‐beta oligomers and less‐toxic fibrils in regions known to harbor familial Alzheimer's mutations. Angew Chem Int Ed 53: 6888–6892
    1. Selkoe DJ, Hardy J (2016) The amyloid hypothesis of Alzheimer's disease at 25 years. EMBO Mol Med 8: 595–608
    1. Sevigny J, Chiao P, Bussière T, Weinreb PH, Williams L, Maier M, Dunstan R, Salloway S, Chen T, Ling Y et al (2016) The antibody aducanumab reduces Aβ plaques in Alzheimer's disease. Nature 537: 50–56
    1. Shankar GM, Bloodgood BL, Townsend M, Walsh DM, Selkoe DJ, Sabatini BL (2007) Natural oligomers of the Alzheimer amyloid‐beta protein induce reversible synapse loss by modulating an NMDA‐type glutamate receptor‐dependent signaling pathway. J Neurosci 27: 2866–2875
    1. Spires TL, Hyman BT (2004) Neuronal structure is altered by amyloid plaques. Rev Neurosci 15: 267–278
    1. Spruijt CG, Gnerlich F, Smits AH, Pfaffeneder T, Jansen PW, Bauer C, Münzel M, Wagner M, Müller M, Khan F et al (2013) Dynamic readers for 5‐(hydroxy)methylcytosine and its oxidized derivatives. Cell 152: 1146–1159
    1. Sun A, Nguyen XV, Bing G (2002) Comparative analysis of an improved thioflavin‐s stain, Gallyas silver stain, and immunohistochemistry for neurofibrillary tangle demonstration on the same sections. J Histochem Cytochem 50: 463–472
    1. Tanzi RE (2005) The synaptic Abeta hypothesis of Alzheimer disease. Nat Neurosci 8: 977–979
    1. Umehara T, Nakamura Y, Jang MK, Nakano K, Tanaka A, Ozato K, Padmanabhan B, Yokoyama S (2010) Structural basis for acetylated histone H4 recognition by the human BRD2 bromodomain. J Biol Chem 285: 7610–7618
    1. Vassar R, Kuhn PH, Haass C, Kennedy ME, Rajendran L, Wong PC, Lichtenthaler SF (2014) Function, therapeutic potential and cell biology of BACE proteases: current status and future prospects. J Neurochem 130: 4–28
    1. Wagner J, Ryazanov S, Leonov A, Levin J, Shi S, Schmidt F, Prix C, Pan‐Montojo F, Bertsch U, Mitteregger‐Kretzschmar G et al (2013) Anle138b: a novel oligomer modulator for disease‐modifying therapy of neurodegenerative diseases such as prion and Parkinson's disease. Acta Neuropathol 125: 795–813
    1. Wagner J, Krauss S, Shi S, Ryazanov S, Steffen J, Miklitz C, Leonov A, Kleinknecht A, Göricke B, Weishaupt JH et al (2015) Reducing tau aggregates with anle138b delays disease progression in a mouse model of tauopathies. Acta Neuropathol 130: 619–631
    1. White SH (1972) Analysis of the torus surrounding planar lipid bilayer membranes. Biophys J 12: 432–445

Source: PubMed

3
订阅