PEPTIDES AND METHODS FOR TREATING NEURODEGENERATIVE DISORDERS

Information

  • Patent Application
  • 20190160146
  • Publication Number
    20190160146
  • Date Filed
    May 12, 2017
    7 years ago
  • Date Published
    May 30, 2019
    5 years ago
Abstract
Disclosed herein are compositions and methods for treating and preventing neurodegenerative diseases, such as Alzheimer's disease. In some embodiments, the composition comprises a peptide that disrupts the binding between PTPσ and APP, preventing β-amyloidogenic processing of APP without affecting other major substrates of β- and Υ-secretases. Alternatively, in some embodiments, an antibody or a fragment of an antibody against PTPσ or APP may be used to disrupt the binding between PTPσ and APP. In some embodiments, the composition comprises compounds or enzymes, which restore perineuronal balance of PTPσ ligands CS and HS, thereby preventing abnormally increased β-amyloidogenic processing of APP. Compositions and methods disclosed herein can be used in combination to treat and prevent neurodegenerative diseases.
Description
BACKGROUND

Alzheimer's disease (AD) is the most common form of dementia, and its risk accelerates after age 65. With a rapidly expanding aging population, AD is projected to become an overwhelming medical burden to the world.


A definitive pathological hallmark of Alzheimer's disease (AD) is the progressive aggregation of β-amyloid (Aβ) peptides in the brain, a process also known as β-amyloidosis, which is often accompanied by neuroinflammation and formation of neurofibrillary tangles containing Tau, a microtubule binding protein1.


Evidence from human genetic studies showed that overproduction of Aβ due to gene mutations inevitably inflicts cascades of cytotoxic events, ultimately leading to neurodegeneration and decay of brain functions. Cerebral accumulation of Aβ peptides, especially in their soluble forms, is therefore recognized as a key culprit in the development of AD1. In the brain, Aβ peptides mainly derive from sequential cleavage of neuronal Amyloid Precursor Protein (APP) by the β- and γ-secretases. However, despite decades of research, molecular regulation of the amyloidogenic secretase activities remains poorly understood, hindering the design of therapeutics to specifically target the APP amyloidogenic pathway.


Pharmacological inhibition of the β- and γ-secretase activities, although effective in suppressing Aβ production, interferes with physiological function of the secretases on their other substrates. Such intervention strategies therefore are often innately associated with untoward side effects, which have led to several failed clinical trials in the past2-4. To date, no therapeutic regimen is available to prevent the onset of AD or curtail its progression.


Besides Aβ, Tau is another biomarker that has been intensively studied in AD. Cognitive decline in patients sometimes correlates better with Tau pathology than with Aβ burden5,6 Overwhelming evidence also substantiated that malfunction of Tau contributes to synaptic loss and neuronal deterioration7.


In addition to AD, many other neurodegenerative diseases also involves Aβ or Tau pathologies, and there is no disease modifying therapy available for any of these debilitating diseases.


SUMMARY

Disclosed herein are peptides, compositions, and methods to treat and prevent neurodegenerative diseases that involve β-amyloid pathologies and/or Tau pathologies, including but not limited to Alzheimer's disease, Lewy body dementia, frontotemporal dementia, cerebral amyloid angiopathy, primary age-related tauopathy, chronic traumatic encephalopathy, Parkinson's disease, postencephalitic parkinsonism, Huntington's disease, amyolateral sclerosis, Pick's disease, progressive supranuclear palsy, corticobasal degeneration, Lytico-Bodig disease, ganglioglioma and gangliocytoma, subacute sclerosing panencephalitis, Hallervorden-Spatz disease, and/or Creutzfeldt-Jakob disease.


These peptides, compositions, and methods may also be used to prevent these neurodegenerative diseases in at-risk subjects, such as people with Down syndrome and those who have suffered from brain injuries or cerebral ischemia, as well as the aging population.


In some embodiments, the disclosed peptides, compositions, and methods disrupt the binding between Protein Tyrosine Phosphatase sigma (PTPσ) and APP, preventing β-amyloidogenic processing of APP as well as Tau aggregation.


In some embodiments, the disclosed compositions and methods restore the physiological balance of two classes of PTPσ ligands in the brain microenvironment, namely the chondroitin sulfates (CS) and heparin or its analog heparan sulfates (HS), and thereby prevent abnormally increased β-amyloidogenic processing of APP.


Unlike the anti-Aβ antibodies in current clinical trials that passively clear β-amyloid, the therapeutic strategy disclosed herein inhibits the process upstream of β-amyloid production. Unlike the β- and γ-secretase inhibitors in current clinical trials, the therapeutic strategy disclosed herein inhibits β-amyloid production without affecting other major substrates of these secretases. Therefore the strategy disclosed herein may be more effective with fewer side effects compared to the most advanced AD drug candidates in clinical trials.


Disclosed herein is a peptide for treating or preventing the aforementioned neurodegenerative disorders, the peptide comprising a decoy fragment of APP, a decoy fragment of PTPσ, or a combination thereof. In some embodiments, the decoy fragment of APP is a peptide comprising at least 5 consecutive amino acids of SEQ ID NO:1. In some embodiments, the decoy fragment of APP is a peptide comprising at least 10 consecutive amino acids of SEQ ID NO:1. For example, the decoy fragment of APP can comprise an amino acid sequence selected from the group consisting of SEQ ID NO:88, SEQ ID NO:91, SEQ ID NO:101, SEQ ID NO:112, SEQ ID NO:139, SEQ ID NO:151, SEQ ID NO:157, SEQ ID NO:251, SEQ ID NO:897. In some embodiments, the decoy fragment of PTPσ is a peptide comprising at least 4 consecutive amino acids of SEQ ID NO:442. For example, the decoy fragment of PTPσ can comprises the amino acid sequence SEQ ID NO:655, SEQ ID NO:769, SEQ ID NO:898, or SEQ ID NO:899. In some embodiments, the peptide further comprises a blood brain barrier penetrating sequence. For example, the blood brain barrier penetrating sequence comprises amino acid sequence SEQ ID NO: 880, SEQ ID NO: 883, SEQ ID NO: 888, SEQ ID NO: 894, SEQ ID NO: 895, SEQ ID NO: 896.


Also disclosed is a method that restores the physiological molecular CS/HS balance that may be used to treat and prevent aforementioned neurodegenerative diseases. In some embodiments, administering HS, or its analog heparin, or their mimetics modified to reduce anti-coagulant effect, with a saccharide chain length of 17, 18, 19, 20, 21, 22, 23, 24 units or longer, could assist in restoring the CS/HS balance. In some embodiments, the physiological molecular CS/HS balance is restored by administering enzymes that digest CS (such as Chondroitinase ABC, also known as ChABC) or prevent HS degradation (such as Heparanase inhibitors PI-88, OGT 2115, or PG545). Alternatively or in addition, agents that mimic the HS/heparin effect of PTPσ clustering8, such as multivalent antibodies, could be administered.


Also disclosed is a method of treating a neurodegenerative disorder in a subject, the method comprising administering to the subject an aforementioned composition or combination of compositions. In some embodiments, the neurodegenerative disease is selected from the group consisting of Alzheimer's Disease, Lewy body dementia, frontotemporal dementia, cerebral amyloid angiopathy, primary age-related tauopathy, chronic traumatic encephalopathy, Parkinson's disease, postencephalitic parkinsonism, Huntington's disease, amyolateral sclerosis, Pick's disease, progressive supranuclear palsy, corticobasal degeneration, Lytico-Bodig disease, ganglioglioma and gangliocytoma, subacute sclerosing panencephalitis, Hallervorden-Spatz disease, and/or Creutzfeldt-Jakob disease. In some embodiments, subjects are selected from at-risk populations, such as the aging population, people with Down syndrome, and those suffered from brain injuries or cerebral ischemia, to prevent subsequent onset of neurodegenerative diseases.


Also disclosed is a method of screening for candidate compounds that slow, stop, reverse, or prevent neurodegeneration. In some embodiments, the method comprises providing a sample comprising APP and PTPσ in an environment permissive for APP-PTPσ binding, contacting the sample with a candidate compound, and assaying the sample for APP-PTPσ binding, wherein a decrease in APP-PTPσ binding compared to control values is an indication that the candidate agent is effective to slow, stop, reverse, or prevent neurodegeneration. In some embodiments, the method comprises contacting/incubating a candidate compound with cell membrane preparations extracted from fresh rodent brain homogenates, wherein a decrease in APP β- and/or γ-cleavage products is an indication that the candidate agent has the potential to slow, stop, reverse, or prevent neurodegeneration.


The details of one or more embodiments of the invention are set forth in the accompanying drawings and the description below. Other features, objects, and advantages of the invention will be apparent from the description and drawings, and from the claims.





DESCRIPTION OF DRAWINGS


FIGS. 1A-1I. PTPσ is an APP binding partner in the brain. a-f, Colocalization of PTPσ (a, green) and APP (b, red) in hippocampal CA1 neurons of adult rat is shown by confocal imaging. Nuclei of CA1 neurons are stained with DAPI (c, blue). d, Merge of three channels. Scale bar, 50 μm. e, Zoom-in image of the soma layer in d. Arrows, intensive colocalization of PTPσ and APP in the initial segments of apical dendrites; arrow heads, punctates of colocalization in the perinuclear regions. Scale bar, 20 μm. f, Zoom-in image of the very fine grained punctates in the axonal compartment in d. Arrows points to the colocalization of PTPσ and APP in axons projecting perpendicular to the focal plane. Scale bar, 10 μm. g, Schematic diagram of PTPσ expressed on cell surface as a two-subunit complex. PTPσ is post-translationally processed into an extracellular domain (ECD) and a transmembrane-intracellular domain (ICD). These two subunits associate with each other through noncovalent bond. Ig-like, immunoglobulin-like domains; fibronectin III-like domains; D1 and D2, two phosphatase domains. h, i, Co-immunoprecipitation (co-IP) of PTPσ and APP from mouse forebrain lysates. Left panels, expression of PTPσ and APP in mouse forebrains. Right panels, IP using an antibody specific for the C-terminus (C-term) of APP. Full length APP (APP FL) is detected by anti-APP C-term antibody. h, PTPσ co-IP with APP from forebrain lysates of wild type but not PTPσ-deficient mice (Balb/c background), detected by an antibody against PTPσ-ECD. i, PTPσ co-IP with APP from forebrain lysates of wild type but not APP knockout mice (B6 background), detected by an antibody against PTPσ-ICD. Dotted lines in i indicate lanes on the same western blot exposure that were moved adjacent to each other. Images shown are representatives of at least three independent experiments using mice between ages of 1 month to 2 years.



FIGS. 2A-2C. Molecular complex of PTPσ and APP in brains of various rodent species. a, b, Co-immunoprecipitation using an anti-APP antibody specific for amino acid residues 1-16 of mouse Aβ (clone M3.2). PTPσ and APP binding interaction is detected in forebrains of Balb/c (a) and B6 (b) mice. c, PTPσ co-immunoprecipitates with APP from rat forebrain lysates using an antibody specific for the C-terminus of APP. Images shown are representatives of at least three independent experiments using different animals.



FIGS. 3A-3I. Genetic depletion of PTPσ reduces β-amyloidogenic products of APP. a, Schematic diagram showing amyloidogenic processing of APP by the β- and γ-secretases. Full length APP (APP FL) is cleaved by β-secretase into soluble N-terminal (sAPPβ) and C-terminal (CTFβ) fragments. APP CTFβ can be further processed by γ-secretase into a C-terminal intracellular domain (AICD) and an Aβ peptide. Aggregation of Aβ is a definitive pathology hallmark of AD. b, PTPσ deficiency reduces the level of an APP CTF at about 15 KD in mouse forebrain lysates, without affecting the expression of APP FL. Antibody against the C-terminus of APP recognizes APP FL and CTFs of both mouse and human origins. c and d, The 15 KD APP CTF is identified as CTFβ by immunoprecipitation (IP) followed with western blot analysis, using a pair of antibodies as marked in the diagram (a). Antibodies against amino acids 1-16 of Aβ (anti-Aβ 1-16) detect CTFβ but not CTFα, as the epitope is absent in CTFα. c, Mouse endogenous CTFβ level is reduced in PTPσ-deficient mouse brains. 4 repeated experiments were quantified by densitometry. d, Human transgenic CTFβ level is reduced in PTPσ-deficient mouse brains harboring human APP-SwDI transgene. 6 repeated experiments were quantified by densitometry. Within each experiment in both c and d, the value from PTPσ deficient sample was normalized to that from the sample with wild type PTPσ. e and f, PTPσ deficiency reduces the levels of Aβ40 (e) and Aβ42 (f) in TgAPP-SwDI mice as measured by ELISA assays. n=12 for each group. The mean values from PTPσ deficient samples was normalized to that from the samples with wild type PTPσ. g and h, Aβ deposition in the hippocampus of 10-month old TgAPP-SwDI mice. Images shown are representatives of 5 pairs of age- and sex-matched mice between 9- to 11-month old. Aβ (green) is detected by immunofluore scent staining using anti-Aβ antibodies clone 6E10 (g) and clone 4G8 (h). DAPI staining is shown in blue. PTPσ deficiency significantly decreases Aβ burden in the brains of TgAPP-SwDI mice. h, Upper panels, the stratum oriens layer between dorsal subiculum (DS) and CA1 (also shown with arrows in g); middle panels, oriens layer between CA1 and CA2; lower panels, the hilus of dentate gyrus (DG, also shown with arrow heads in g). Left column, control staining without primary antibody (no 1° Ab). No Aβ signal is detected in non-transgenic mice (data not shown). Scale bars, 500 μm in g and 100 μm in h. i, Genetic depletion of PTPσ suppresses the progression of Aβ pathology in TgAPP-SwDI mice. ImageJ quantification of Aβ immunofluorescent staining (with 6E10) in DG hilus from 9- and 16-month old TgAPP-SwDI mice. n=3 for each group. Total integrated density of Aβ in DG hilus was normalized to the area size of the hilus to yield the average intensity as show in the bar graph. Mean value of each group was normalized to that of 16 month old TgAPP-SwDI mice expressing wild type PTPσ. All p values, Student's t test, 2-tailed. Error bars, SEM.



FIGS. 4A-4F. Genetic depletion of PTPσ reduces β-amyloidogenic products of APP. a and b, Antibody against the C-terminus of APP recognizes full length (FL) and C-terminal fragments (CTFs) of both mouse and human APP. PTPσ deficiency does not affect the expression level of APP FL (a), but reduces the level of an APP CTF at about 15 KD in mouse forebrain lysates (b). Images shown are representatives of at least three independent experiments. c, Human CTFβ in the forebrains of APP-SwInd transgenic mice is identified using the method as described in FIG. 2d. CTFβ is immunoprecipitated by an antibody against the C-terminus of APP and detected by western blot analysis using an antibody against amino acids 1-16 of human Aβ (6E10), which reacts with CTFβ but not CTFα (regions of antibody epitopes are shown in FIG. 2a). d, Densitometry quantification of experiments as shown in panel c repeated with 5 pairs of mice. For each experiment, the value from PTPσ deficient sample was normalized to the value from the sample with wild type PTPσ. e, Representative images of Aβ immunofluorescent staining (with 6E10) in the hippocampus of 15-month old TgAPP-SwInd mice. Arrows point to Aβ deposits. Scale bars, 50 μm. f, Aβ immunofluorescent staining in the hippocampus of 15-month old TgAPP-SwInd mice, as shown in panel e, was quantified using ImageJ. APP-SwInd(+)PTPσ(+/+), n=7; APP-SwInd(+)PTPσ(−/−), n=8. The mean value of APP-SwInd(+)PTPσ(−/−) samples was normalized to that of APP-SwInd(+)PTPσ(+/+) samples. All error bars, SEM. All p values, Student's t test, 2-tailed.



FIGS. 5A-5C. Lower affinity between BACE1 and APP in PTPσ-deficient brains. a, Co-immunoprecipitation experiments show nearly equal BACE1-APP association in wild type and PTPσ-deficient mouse brains under mild detergent condition (1% NP40). However, in PTPσ-deficient brains, BACE1-APP association detected by co-immunoprecipitation is more vulnerable to increased detergent stringency as compared to that in wild type brains. Panels of blots show full length APP (APP FL) pulled down with an anti-BACE1 antibody from mouse forebrain lysates. NP40, Nonidet P-40, non-ionic detergent. SDS, Sodium dodecyl sulfate, ionic detergent. b, Co-immunoprecipitation under buffer condition with 1% NP40 and 0.3% SDS, as shown in the middle panel of a, were repeated with three pair of mice. Each experiment was quantified by densitometry, and the value from PTPσ-deficient sample was calculated as a percentage of that from the wild type sample (also shown as orange points in c). Error bar, SEM. p value, Student's t test, 2-tailed. c, Co-immunoprecipitation experiments were repeated under each detergent condition. The percentage values shown in dots are derived using the same method as in b. Bars represent means. Increasingly stringent buffer conditions manifest a lower BACE1-APP affinity in PTPσ-deficient brains. p value and R2, linear regression.



FIGS. 6A-6F. PTPσ does not generically modulate b- and g-secretases. Neither expression levels of the secretases or their activities on other major substrates are affected by PTPσ depletion. Mouse forebrain lysates with or without PTPσ were analyzed by western blot. a and b, PTPσ deficiency does not change expression level of BACE1 (a) or γ-secretase subunits (b). Presenilin1 and 2 (PS1/2) are the catalytic subunits of γ-secretase, which are processed into N-terminal and C-terminal fragments (NTF and CTF) in their mature forms. Nicastrin, Presenilin Enhancer 2 (PEN2), and APH1 are other essential subunits of γ-secretase. c, PTPσ deficiency does not change the level of Neuregulinl (NGR1) CTFβ, the C-terminal cleavage product by BACE1. NRG1 FL, full length Neuregulinl. d, The level of Notch cleavage product by γ-secretase is not affected by PTPσ deficiency. TMIC, Notch transmembrane/intracellular fragment, which can be cleaved by γ-secretase into a C-terminal intracellular domain NICD (detected by an antibody against Notch C-terminus in the upper panel, and by an antibody specific for γ-secretase cleaved NICD in the lower panel). e, Actin loading control for a and c. f, Actin loading control for b and d. All images shown are representatives of at least three independent experiments. All images shown are representatives of at least three independent experiments using different animals.



FIGS. 7A-7K. PTPσ deficiency attenuates reactive astrogliosis in APP transgenic mice. Expression level of GFAP, a marker of reactive astrocytes, is suppressed in the brains of TgAPP-SwDI mice by PTPσ depletion. Representative images show GFAP (red) and DAPI staining of nuclei (blue) in the brains of 9-month old TgAPP-SwDI mice with or without PTPσ, along with their non-transgenic wild type littermate. a-f, Dentate gyms (DG) of the hippocampus; scale bars, 100 μm. g-j, Primary somatosensory cortex; scale bars, 200 μm. k, ImageJ quantification of GFAP level in DG hilus from TgAPP-SwDI mice aged between 9 to 11 months. APP-SwDI(−)PTPσ(+/+), non-transgenic wild type littermates (expressing PTPσ but not the human APP transgene). Total integrated density of GFAP in DG hilus was normalized to the area size of the hilus to yield average intensity as shown in the bar graph. Mean value of each group was normalized to that of APP-SwDI(−)PTPσ(+/+) mice. APP-SwDI(−)PTPσ(+/+), n=4; APP-SwDI(+)PTPσ(+/+), n=4; APP-SwDI(+)PTPσ(−/−), n=6. All p values, Student's t test, 2-tailed. Error bars, SEM.



FIGS. 8A-8G. PTPσ deficiency protects APP transgenic mice from synaptic loss. Representative images show immunofluorescent staining of presynaptic marker Synaptophysin in the mossy fiber terminal zone of CA3 region. a-f, Synaptophysin, red; DAPI, blue. Scale bars, 100 μm. g, ImageJ quantification of Synaptophysin expression level in CA3 mossy fiber terminal zone from mice aged between 9 to 11 months. Total integrated density of Synaptophysin in CA3 mossy fiber terminal zone was normalized to the area size to yield average intensity as shown in the bar graph. Mean value of each group was normalized to that of wild type APP-SwDI(−)PTPσ(+/+) mice. APP-SwDI(−)PTPν(+/+), n=4; APP-SwDI(+)PTPσ(+/+), n=6; APP-SwDI(+)PTPσ(−/−), n=6. All p values, Student's t test, 2-tailed. Error bars, SEM.



FIGS. 9A-91I. PTPσ deficiency mitigates Tau pathology in TgAPP-SwDI mice. a, Schematic diagram depicting distribution pattern of Tau aggregation (green) detected by immunofluorescent staining using an anti-Tau antibody (Tau-5) against its proline-rich region, in brains of 9 to 11 month-old TgAPP-SwDI transgenic mice. Similar results are seen with Tau-46, an antibody recognizing the C-terminus of Tau (Extended Data FIG. 6). Aggregated Tau is found most prominently in the molecular layer of piriform and entorhinal cortex, and occasionally in hippocampal regions in APP-SwDI(+)PTPσ(+/+) mice. b, PTPσ deficiency diminishes Tau aggregation. Bar graph shows quantification of Tau aggregation in coronal brain sections from 4 pairs of age- and sex-matched APP-SwDI(+)PTPσ(+/+) and APP-SwDI(+)PTPσ(−/−) mice of 9 to 11 month-old. For each pair, the value from APP-SwDI(+)PTPσ(−/−) sample is normalized to the value from APP-SwDI(+)PTPσ(+/+) sample. p value, Student's t test, 2-tailed. Error bar, SEM. c, d, Representative images of many areas with Tau aggregation in APP-SwDI(+)PTPσ(+/+) brains. f, g, Representative images of a few areas with Tau aggregation in age-matched APP-SwDI(+)PTPσ(−/−) brains. c and f, Hippocampal regions. d-h, Piriform cortex. e, Staining of a section adjacent to d, but without primary antibody (no 1° Ab). h, no Tau aggregates are detected in aged-matched non-transgenic wild type littermates (expressing PTPσ but not the human APP transgene). Tau, green; DAPI, blue. Arrows points to Tau aggregates. Scale bars, 50 μm.



FIGS. 10A-10E. PTPσ deficiency mitigates Tau pathology in TgAPP-SwInd mice. Tau aggregation (green) is detected by immunofluorescent staining, using an anti-Tau antibody (Tau-5, as in FIG. 5) in the brains of 15 month-old TgAPP-SwInd transgenic mice. Similar results are seen with Tau-46, an antibody recognizing the C-terminus of Tau (Extended Data FIG. 6). Aggregated Tau is found most prominently in the molecular layer of the entorhrinal (a, b) and piriform cortex (c, d), and occasionally in the hippocampal regions (images not shown). e, PTPσ deficiency diminishes Tau aggregation as quantified in coronal brain sections from 15 month-old APP-SwInd(+)PTPσ(+/+) (n=7) and APP-SwInd(+)PTPσ(−/−) mice (n=8). The mean value of APP-SwInd(+)PTPσ(−/−) samples is normalized to that of APP-SwInd(+)PTPσ(+/+). p value, Student's t test, 2-tailed. Error bars, SEM. Tau, green; DAPI, blue. Arrows points to Tau aggregates. Scale bars, 50 μm.



FIGS. 11A-11J. Morphology of Tau aggregates found in APP transgenic brains. a-h, Tau aggregation (green) is detected by immunofluorescent staining, using an anti-Tau antibody (Tau-5) against the proline-rich domain of Tau (same as in FIG. 5 and Extended Data FIG. 5). Tau aggregates in TgAPP-SwDI and TgAPP-SwInd brains show similar morphologies. a-f, Many of the Tau aggregates are found in punctate shapes, likely as part of cell debris, in areas that are free of nuclei staining. g, h, Occasionally the aggregates are found in fibrillary structures, probably in degenerated cells before disassembling. i, An additional anti-Tau antibody (Tau-46), which recognizes the C-terminus of Tau, detects Tau aggregation in the same pattern as Tau-5. j, Image of staining without primary antibody at the same location of the Tau aggregates in the section adjacent to i. Both these antibodies recognize Tau regardless of its phosphorylation status. Tau, green; DAPI, blue. All scale bars, 20 μm.



FIG. 12. Tau expression is not affected by PTPσ or human APP transgenes. Upper panel, total Tau level in brain homogenates. Lower panel, Actin as loading control. Tau protein expression level is not changed by genetic depletion of PTPσ or expression of mutated human APP transgenes. All mice are older than 1 year, and mice in each pair are age- and sex matched. Images shown are representatives of three independent experiments.



FIGS. 13A-13C. PTPσ deficiency rescues behavioral deficits in TgAPP-SwDI mice. a, In the Y-maze assay, performance of spatial navigation is scored by the percentage of spontaneous alternations among total arm entries. Values are normalized to that of non-transgenic wild type APP-SwDI(−)PTPσ(+/+) mice within the colony. Compared to non-transgenic wild type mice, APP-SwDI(+)PTPσ(+/+) mice show deficit of short-term spatial memory, which is rescued by genetic depletion of PTPσ in APP-SwDI(+)PTPσ(−/−) mice. APP-SwDI(−)PTPσ(+/+), n=23 (18 females and 5 males); APP-SwDI(+)PTPσ(+/+), n=52 (30 females and 22 males); APP-SwDI(+)PTPσ(−/−), n=35 (22 females and 13 males). Ages of all genotype groups are similarly distributed between 4 and 11 months. b, c, Novel object test. NO, novel object. FO, familiar object. Attention to NO is measured by the ratio of NO exploration to total object exploration (NO+FO) in terms of exploration time (b) and visiting frequency (c). Values are normalized to that of non-transgenic wild type mice. APP-SwDI(+)PTPσ(+/+) mice showed decreased interest in NO compared to wild type APP-SwDI(−)PTPσ(+/+) mice. The deficit is reversed by PTPσ depletion in APP-SwDI(+)PTPσ(−/−) mice. APP-SwDI(−)PTPσ(+/+), n=28 (19 females and 9 males); APP-SwDI(+)PTPσ(+/+), n=46 (32 females and 14 males); APP-SwDI(+)PTPσ(−/−), n=29 (21 females and 8 males). Ages of all groups are similarly distributed between 4 and 11 months. All p values, Student's t test, 2-tailed. Error bars, SEM.



FIG. 14. PTPσ deficiency restores short-term spatial memory in TgAPP-SwDI mice. In the Y-maze assay, performance of spatial navigation is scored by the percentage of spontaneous alternations among total arm entries. The raw values shown here are before normalization in FIG. 6a. Compared to non-transgenic wild type APP-SwDI(−)PTPσ(+/+)mice, APP-SwDI(+)PTPσ(+/+) mice show deficit of short-term spatial memory, which is rescued by genetic depletion of PTPσ. APP-SwDI(−)PTPσ(+/+), n=23 (18 females and 5 males); APP-SwDI(+)PTPσ(+/+), n=52 (30 females and 22 males); APP-SwDI(+)PTPσ(−/−), n=35 (22 females and 13 males). Ages of all genotype groups are similarly distributed between 4 and 11 months. All p values, Student's t test, 2-tailed. Error bars, SEM.



FIGS. 15A-15D. PTPσ deficiency enhances novelty exploration by TgAPP-SwDI mice. NO, novel object. FO, familiar object. a and b, In novel object test, NO preference is measured by the ratio between NO and FO exploration, where NO/FO>1 indicates preference for NO. c and d, Attention to NO is additionally measured by the discrimination index, NO/(NO+FO), the ratio of NO exploration to total object exploration (NO+FO). The raw values shown here in c and d are before normalization in FIGS. 6b and c. Mice of this colony show a low baseline of the NO/(NO+FO) discrimination index, likely inherited from their parental Balb/c line. For non-transgenic wild type APP-SwDI(−)PTPσ(+/+) mice, the discrimination index is slightly above 0.5 (chance value), similar to what was previously reported for the Balb/c wild type mice27. Thus, a sole measurement of the discrimination index may not reveal the preference for NO as does the NO/FO ratio. Although not as sensitive in measuring object preference, the NO/(NO+FO) index is most commonly used as it provides a normalization of the NO exploration to total object exploration activity. While each has its own advantage and shortcoming, both NO/FO and NO/NO+FO measurements consistently show that the expression of TgAPP-SwDI gene leads to a deficit in attention to the NO, whereas genetic depletion of PTPσ restores novelty exploration to a level close to that of non-transgenic wild type mice. a and c, measurements in terms of exploration time. b and d, measurements in terms of visiting frequency. APP-SwDI(−)PTPσ(+/+), n=28 (19 females and 9 males); APP-SwDI(+)PTPσ(+/+), n=46 (32 females and 14 males); APP-SwDI(+)PTPσ(−/−), n=29 (21 females and 8 males). Ages of all groups are similarly distributed between 4 and 11 months. All p values, Student's t test, 2-tailed. Error bars, SEM.



FIGS. 16A-16C. PTPσ deficiency improves behavioral performance of TgAPP-SwInd mice. a, Performance of spatial navigation is scored by the percentage of spontaneous alternations among total arm entries in the Y-maze assay. Compared to APP-SwInd(+)PTPσ(+/+) mice, APP-SwInd(+)PTPσ(−/−) mice showed improved short-term spatial memory. APP-SwInd(+)PTPσ(+/+), n=40 (20 females and 20 males); APP-SwInd(+)PTPσ(−/−), n=18 (9 females and 9 males). Ages of both genotype groups are similarly distributed between 4 and 11 months. b, c, Novel object test. NO, novel object. FO, familiar object. NO preference is measured by the ratio of NO exploration time to total object exploration time (b) and the ratio of NO exploration time to FO exploration time (c). PTPσ depletion significantly improves novelty preference in these transgenic mice. APP-SwInd(+)PTPσ(+/+), n=43 (21 females and 22 males); APP-SwInd(+)PTPσ(−/−), n=24 (10 females and 14 males). Ages of both groups are similarly distributed between 5 and 15 months. All p values, Student's t test, 2-tailed. Error bars, SEM.



FIG. 17. CS and HS regulate β-cleavage of APP in opposite manners. Membrane preparations from fresh mouse brain homogenates are incubated with CS18 (chondroitin sulfate of 18 oligosaccharides) or HS17 (heparan sulfate analog, heparin fragment of 17 oligosaccharides) at 37 C.° for 30 min. Levels of APP β-cleavage product (CTFβ) as detected by Western blot analysis are enhanced by CS18 treatment but diminished by HS17 treatment. FL APP, full length APP. Control, no treatment.



FIGS. 18A and 18B. TBI enhances PTPσ-APP binding and β-cleavage of APP. a, Co-immunoprecipitation of PTPσ with APP showed increased PTPσ-APP binding in after TBI in rat. b, Level of APP β-cleavage product (CTFβ) is enhanced in correlation with increased PTPσ-APP binding. Similar results are found using in mouse TBI brains.



FIG. 19 Heparin fragment of 17 oligosaccharides inhibits APP-PTPσ binding. Recombinant human APP fragment binding to PTPσ is detected by kinetic ELISA assay. Heparin fragment of 17 oligosaccharides (heparan sulfate analog) effectively disrupts APP-PTPσ binding when included in the binding assay. APP fragment used here corresponds to SEQ ID NO:1, which is the region between E1 and E2 domains. PTPσ fragment used here includes its IG1 and IG2 domains.



FIG. 20 Ligand binding site of PTPσ IG1 domain interacts with APP. Binding of human APP fragment (SEQ ID NO:1) with various PTPσ fragments is measured by kinetic ELISA assay. APP fragment corresponds to SEQ ID NO:1, which is a region between E1 and E2 domains. PTPσ fragments used here include IG1,2 (containing IG1 and IG2 domains), ΔLysIG1,2 (containing IG1 and IG2 domains, with lysine 67, 68, 70, 71 mutated to alanine), IG1-FN1 (containing IG1, IG2, IG3 and FN1 domains), ECD (full extracellular domain of PTPσ containing all 3 IG domains and 4 FN domains). Value shown are mean±SEM, n=3 for each group. ***, p≤0.001, Student t test, comparison with the IG1,2.





DETAILED DESCRIPTION

Experimental results in Example 1 show that neuronal receptor PTPσ mediates both (3-amyloid and Tau pathogenesis in two mouse models. In the brain, PTPσ binds to APP. Depletion of PTPσ reduces the affinity between APP and β-secretase, diminishing APP proteolytic products by β- and γ-cleavage without affecting other major substrates of the secretases, suggesting a specificity of β-amyloidogenic regulation. In human APP transgenic mice during aging the progression of β-amyloidosis, Tau aggregation, neuroinflammation, synaptic loss, as well as behavioral deficits, all show unambiguous dependency on the expression of PTPσ. Additionally, the aggregates of endogenous Tau are found in a distribution pattern similar to that of early stage neurofibrillary tangles in Alzheimer brains. Together, these findings unveil a gatekeeping role of PTPσ upstream of the degenerative pathogenesis, indicating a potential for this neuronal receptor as a drug target for Alzheimer's disease.


Experimental results in Example 2 show that two classes of PTPσ ligands in the brain microenvironment, CS and HS, regulate APP amyloidogenic processing in opposite manners. CS increases APP β-cleavage products, whereas HS decreases APP β-cleavage products. Because CS and HS compete to interact with receptor PTPσ yet lead to opposite signaling and neuronal responses, the ratio of perineuronal CS and HS is therefore crucial for the downstream effects of PTPσ and maintaining the health of the brain.


Experimental results in Example 3 further define that the binding between APP and PTPσ is mediated by a fragment on APP between its E1 and E2 domain and the IG1 domain of PTPσ.


The findings that PTPσ plays a pivotal role in the development of β-amyloid and Tau pathologies indicate that peptides, compositions, and methods disclosed herein may be suitable to treat and prevent neurodegenerative diseases that involve β-amyloid pathologies and/or Tau pathologies, including but not limited to Alzheimer's disease, Lewy body dementia, frontotemporal dementia, cerebral amyloid angiopathy, primary age-related tauopathy, chronic traumatic encephalopathy, Parkinson's disease, postencephalitic parkinsonism, Huntington's disease, amyolateral sclerosis, Pick's disease, progressive supranuclear palsy, corticobasal degeneration, Lytico-Bodig disease, ganglioglioma and gangliocytoma, subacute sclerosing panencephalitis, Hallervorden-Spatz disease, and/or Creutzfeldt-Jakob disease.


Additionally, these peptides, compositions, and methods may also be used to prevent these neurodegenerative diseases in at-risk populations, such as subjects with Down syndrome and those suffered from brain injuries or cerebral ischemia, as well as the aging population.


Definitions

As used in the specification and claims, the singular form “a,” “an,” and “the” include plural references unless the context clearly dictates otherwise. For example, the term “a cell” includes a plurality of cells, including mixtures thereof.


The terms “about” and “approximately” are defined as being “close to” as understood by one of ordinary skill in the art. In one non-limiting embodiment the terms are defined to be within 10%. In another non-limiting embodiment, the terms are defined to be within 5%. In still another non-limiting embodiment, the terms are defined to be within 1%.


The terms “protein,” “peptide,” and “polypeptide” are used interchangeably to refer to a natural or synthetic molecule comprising two or more amino acids linked by the carboxyl group of one amino acid to the alpha amino group of another. The term “protein” includes amino acids joined to each other by peptide bonds or modified peptide bonds, e.g., peptide isosteres, etc., and can contain modified amino acids other than the 20 gene-encoded amino acids. The polypeptides can be modified by either natural processes, such as post-translational processing, or by chemical modification techniques which are well known in the art. The term also includes peptidomimetics and cyclic peptides.


As used herein, “peptidomimetic” means a mimetic of a peptide which includes some alteration of the normal peptide chemistry. Peptidomimetics typically enhance some property of the original peptide, such as increase stability, increased efficacy, enhanced delivery, increased half life, etc. Methods of making peptidomimetics based upon a known polypeptide sequence is described, for example, in U.S. Pat. Nos. 5,631,280; 5,612,895; and 5,579,250. Use of peptidomimetics can involve the incorporation of a non-amino acid residue with non-amide linkages at a given position. One embodiment of the present invention is a peptidomimetic wherein the compound has a bond, a peptide backbone or an amino acid component replaced with a suitable mimic. Some non-limiting examples of unnatural amino acids which may be suitable amino acid mimics include β-alanine, L-α-amino butyric acid, L-γ-amino butyric acid, L-α-amino isobutyric acid, L-ε-amino caproic acid, 7-amino heptanoic acid, L-aspartic acid, L-glutamic acid, N-ε-Boc-N-α-CBZ-L-lysine, N-ε-Boc-N-α-Fmoc-L-lysine, L-methionine sulfone, L-norleucine, L-norvaline, N-α-Boc-N-δCBZ-L-ornithine, N-δ-Boc-N-α-CBZ-L-ornithine, Boc-p-nitro-L-phenylalanine, Boc-hydroxyproline, and Boc-L-thioproline.


A “fusion protein” refers to a polypeptide formed by the joining of two or more polypeptides through a peptide bond formed between the amino terminus of one polypeptide and the carboxyl terminus of another polypeptide. The fusion protein can be formed by the chemical coupling of the constituent polypeptides or it can be expressed as a single polypeptide from nucleic acid sequence encoding the single contiguous fusion protein. A single chain fusion protein is a fusion protein having a single contiguous polypeptide backbone. Fusion proteins can be prepared using conventional techniques in molecular biology to join the two genes in frame into a single nucleic acid, and then expressing the nucleic acid in an appropriate host cell under conditions in which the fusion protein is produced.


As used herein, protein “binding” is the binding of one protein to another. The binding may comprise covalent bonds, protein cross-linking, and/or non-covalent interactions such as hydrophobic interactions, ionic interactions, or hydrogen bonds.


The term “protein domain” refers to a portion of a protein, portions of a protein, or an entire protein showing structural integrity; this determination may be based on amino acid composition of a portion of a protein, portions of a protein, or the entire protein.


“Amyloid precursor protein” (APP) is an integral membrane protein expressed in many tissues and concentrated in the synapses of neurons. It has been implicated as a regulator of synapse formation, neural plasticity and iron export. APP is cleaved by beta secretase and gamma secretase to yield Aβ. Amyloid beta (Aβ) denotes peptides of 36-43 amino acids that are involved in Alzheimer's disease as the main component of the amyloid plaques found in the brains of Alzheimer patients. Aβ molecules cleaved from APP can aggregate to form flexible soluble oligomers which may exist in various forms. Certain misfolded oligomers (known as “seeds”) can induce other Aβ molecules to also take the misfolded oligomeric foam, leading to a chain reaction and buildup of amyloid plaques. The seeds or the resulting amyloid plaques are toxic to cells in the brain.


“Protein tyrosine phosphatases” or “receptor protein tyrosine phosphatases” (PTPs) are a group of enzymes that remove phosphate groups from phosphorylated tyrosine residues on proteins. Protein tyrosine phosphorylation is a common post-translational modification that can create novel recognition motifs for protein interactions and cellular localization, affect protein stability, and regulate enzyme activity. As a consequence, maintaining an appropriate level of protein tyrosine phosphorylation is essential for many cellular functions. Tyrosine-specific protein phosphatases catalyze the removal of a phosphate group attached to a tyrosine residue. These enzymes are key regulatory components in many signal transduction pathways (such as the MAP kinase pathway) that underlie cellular functions such as cell cycle control/proliferation, cell death, differentiation, transformation, cell polarity and motility, synaptic plasticity, etc.


The term “subject” refers to any individual who is the target of administration or treatment. The subject can be a vertebrate, for example, a mammal. Thus, the subject can be a human or veterinary patient. The term “patient” refers to a subject under the treatment of a clinician, e.g., physician. An “at-risk” subject is an individual with a higher likelihood of developing a certain disease or condition. An “at-risk” subject may have, for example, received a medical diagnosis associated with the certain disease or condition.


“Tau proteins” (or τ proteins) are proteins that stabilize microtubules. They are abundant in neurons of the central nervous system and are less common elsewhere, but are also expressed at very low levels in CNS astrocytes and oligodendrocytes. Neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, and other tauopathies are associated with tau proteins that have become defective, misfolded, tangled, and no longer stabilize microtubules properly.


The term “protein fragment” refers to a functional portion of a full-length protein. For example, a fragment of APP or PTPσ may be synthesized chemically or biologically for the purposes of disrupting the binding between APP and PTPσ. Such fragments could be used as “decoy” peptides to prevent or diminish the actual APP-PTPσ binding interaction that results in β-cleavage of APP and subsequent Aβ formation.


The phrase “functional fragment” or “analog” or mimetic of a protein or other molecule is a compound having qualitative biological activity in common with a full-length protein or other molecule of its entire structure. A functional fragment of a full-length protein may be isolated and attached to a separate peptide sequence. For example, a functional fragment of a blood-brain barrier penetrating protein may be isolated and attached to the decoy peptide that disrupts APP-PTPσ binding, thereby enabling the hybrid peptide to enter the brain and disrupt APP-PTPσ binding. Another example of a functional fragment is a membrane penetrating fragment, or one that relays an ability to pass the lipophilic barrier of a cell's plasma membrane. An analog of heparin, for example, may be a compound that binds to a heparin binding site.


As used herein, “cyclic peptide” or “cyclopeptide” in general refers to a peptide comprising at least one internal bond attaching nonadjacent amino acids of the peptide, such as when the end amino acids of a linear sequence are attached to form a circular peptide.


The term “antibody” refers to natural or synthetic antibodies that selectively bind a target antigen. The term includes polyclonal and monoclonal antibodies. In addition to intact immunoglobulin molecules, also included in the term “antibodies” are fragments or polymers of those immunoglobulin molecules, and human or humanized versions of immunoglobulin molecules that selectively bind the target antigen.


As used herein, “enzyme” refers to a protein specialized to catalyze or promote a specific metabolic reaction.


“Neurodegenerative disorders” or “neurodegenerative diseases” are conditions marked by the progressive loss of structure or function of neural cells, including death of neurons and glia.


The term “treatment” refers to the medical management of a patient with the intent to cure, ameliorate, stabilize, or prevent a disease, pathological condition, or disorder. This term includes active treatment, that is, treatment directed specifically toward the improvement of a disease, pathological condition, or disorder, and also includes causal treatment, that is, treatment directed toward removal of the cause of the associated disease, pathological condition, or disorder. In addition, this term includes palliative treatment, that is, treatment designed for the relief of symptoms rather than the curing of the disease, pathological condition, or disorder; preventative treatment, that is, treatment directed to minimizing or partially or completely inhibiting the development of the associated disease, pathological condition, or disorder; and supportive treatment, that is, treatment employed to supplement another specific therapy directed toward the improvement of the associated disease, pathological condition, or disorder.


The term “administering” refers to an administration that is intranasal, oral, topical, intravenous, subcutaneous, transcutaneous, transdermal, intramuscular, intra-joint, parenteral, intra-arteriole, intradermal, intraventricular, intracranial, intraperitoneal, intralesional, rectal, vaginal, by inhalation or via an implanted reservoir. The term “parenteral” includes subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional, and intracranial injections or infusion techniques.


The term “pharmaceutically acceptable carrier” means a carrier or excipient that is useful in preparing a pharmaceutical composition that is generally safe and non-toxic, and includes a carrier that is acceptable for veterinary and/or human pharmaceutical use. As used herein, the term “pharmaceutically acceptable carrier” encompasses any of the standard pharmaceutical carriers, such as a phosphate buffered saline solution, water, and emulsions, such as an oil/water or water/oil emulsion, and various types of wetting agents. As used herein, the term “carrier” encompasses any excipient, diluent, filler, salt, buffer, stabilizer, solubilizer, lipid, stabilizer, or other material well known in the art for use in pharmaceutical formulations and as described further below. The pharmaceutical compositions also can include preservatives. A “pharmaceutically acceptable carrier” as used in the specification and claims includes both one and more than one such carrier.


The term “variant” refers to an amino acid or peptide sequence having conservative amino acid substitutions (“conservative variant”), non-conservative amino acid subsitutions (e.g., a degenerate variant), substitutions within the wobble position of each codon (i.e. DNA and RNA) encoding an amino acid, amino acids added to the C-terminus of a peptide, or a peptide having 60%, 70%, 80%, 90%, or 95% homology to a reference sequence.


The term “percent (%) sequence identity” or “homology” is defined as the percentage of nucleotides or amino acids in a candidate sequence that are identical with the nucleotides or amino acids in a reference nucleic acid sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity. Alignment for purposes of determining percent sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN, ALIGN-2 or Megalign (DNASTAR) software. Appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the full-length of the sequences being compared can be determined by known methods.


Compositions


Peptides:

Disclosed herein are peptides for treating and preventing the aforementioned neurodegenerative diseases, such as Alzheimer's disease. In some embodiments, the peptides disrupt the binding between PTPσ and APP, preventing β-amyloidogenic processing of APP without affecting other major substrates of the β- and γ-secretases. The peptide may be a decoy fragment of APP, a decoy fragment of PTPσ, or a combination thereof.


In some embodiments, a decoy peptide could be fabricated from the PTPσ-binding region on APP, which is the fragment between its E1 and E2 domains (SEQ ID NO:1). In some embodiments, a decoy peptide could be fabricated from the APP-binding region on PTPσ, which is its IG1 domain (SEQ ID NO: 442). In some embodiments, a decoy peptide could be fabricated that corresponds to the entire APP E2 domain or a fragment thereof. In some embodiments, a decoy peptide could be fabricated that corresponds to the entire APP E1 domain or a fragment thereof. In some embodiments, a PTPσ peptide is used in combination with an APP peptide.


In some embodiments, the peptide is a fragment of the PTPσ-binding domain of APP. Therefore, in some embodiments, the peptide is a fragment of SEQ ID NO:1, as listed below, which has at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or more amino acids, or a conservative variant thereof.









(SEQ ID NO: 1)







AEESDNVDSADAEEDDSDVWWGGADTDYADGSEDKVVEVAEEEEVAEVEE





EEADDDEDDEDGDEVEEEAEEPYEEATERTTSIATTTTTTTESVEEVVR.






Therefore, in some embodiments, the peptide comprises an amino acid sequence selected from 10 consecutive residues of SEQ ID NO: 1, or from the group consisting of the below:









SEQ ID NO: 2









AEESDNVDSA











SEQ ID NO: 3









EESDNVDSAD











SEQ ID NO: 4









ESDNVDSADA











SEQ ID NO: 5









SDNVDSADAE











SEQ ID NO: 6









DNVDSADAEE











SEQ ID NO: 7









NVDSADAEED











SEQ ID NO: 8









VDSADAEEDD











SEQ ID NO: 9









DSADAEEDDS











SEQ ID NO: 10









SADAEEDDSD











SEQ ID NO: 11









ADAEEDDSDV











SEQ ID NO: 12









DAEEDDSDVW











SEQ ID NO: 13









AEEDDSDVWW











SEQ ID NO: 14









EEDDSDVWWG











SEQ ID NO: 15









EDDSDVWWGG











SEQ ID NO: 16









DDSDVWWGGA











SEQ ID NO: 17









DSDVWWGGAD











SEQ ID NO: 18









SDVWWGGADT











SEQ ID NO: 19









DVWWGGADTD











SEQ ID NO: 20









VWWGGADTDY











SEQ ID NO: 21









WWGGADTDYA











SEQ ID NO: 22









WGGADTDYAD











SEQ ID NO: 23









GGADTDYADG











SEQ ID NO: 24









GADTDYADGS











SEQ ID NO: 25









ADTDYADGSE











SEQ ID NO: 26









DTDYADGSED











SEQ ID NO: 27









TDYADGSEDK











SEQ ID NO: 28









DYADGSEDKV











SEQ ID NO: 29









YADGSEDKVV











SEQ ID NO: 30









ADGSEDKVVE











SEQ ID NO: 31









DGSEDKVVEV











SEQ ID NO: 32









GSEDKVVEVA











SEQ ID NO: 33









SEDKVVEVAE











SEQ ID NO: 34









EDKVVEVAEE











SEQ ID NO: 35









DKVVEVAEEE











SEQ ID NO: 36









KVVEVAEEEE











SEQ ID NO: 37









VVEVAEEEEV











SEQ ID NO: 38









VEVAEEEEVA











SEQ ID NO: 39









EVAEEEEVAE











SEQ ID NO: 40









VAEEEEVAEV











SEQ ID NO: 41









AEEEEVAEVE











SEQ ID NO: 42









EEEEVAEVEE











SEQ ID NO: 43









EEEVAEVEEE











SEQ ID NO: 44









EEVAEVEEEE











SEQ ID NO: 45









EVAEVEEEEA











SEQ ID NO: 46









VAEVEEEEAD











SEQ ID NO: 47









AEVEEEEADD











SEQ ID NO: 48









EVEEEEADDD











SEQ ID NO: 49









VEEEEADDDE











SEQ ID NO: 50









EEEEADDDED











SEQ ID NO: 51









EEEADDDEDD











SEQ ID NO: 52









EEADDDEDDE











SEQ ID NO: 53









EADDDEDDED











SEQ ID NO: 54









ADDDEDDEDG











SEQ ID NO: 55









DDDEDDEDGD











SEQ ID NO: 56









DDEDDEDGDE











SEQ ID NO: 57









DEDDEDGDEV











SEQ ID NO: 58









EDDEDGDEVE











SEQ ID NO: 59









DDEDGDEVEE











SEQ ID NO: 60









DEDGDEVEEE











SEQ ID NO: 61









EDGDEVEEEA











SEQ ID NO: 62









DGDEVEEEAE











SEQ ID NO: 63









GDEVEEEAEE











SEQ ID NO: 64









DEVEEEAEEP











SEQ ID NO: 65









EVEEEAEEPY











SEQ ID NO: 66









VEEEAEEPYE











SEQ ID NO: 67









EEEAEEPYEE











SEQ ID NO: 68









EEAEEPYEEA











SEQ ID NO: 69









EAEEPYEEAT











SEQ ID NO: 70









AEEPYEEATE











SEQ ID NO: 71









EEPYEEATER











SEQ ID NO: 72









EPYEEATERT











SEQ ID NO: 73









PYEEATERTT











SEQ ID NO: 74









YEEATERTTS











SEQ ID NO: 75









EEATERTTSI











SEQ ID NO: 76









EATERTTSIA











SEQ ID NO: 77









ATERTTSIAT











SEQ ID NO: 78









TERTTSIATT











SEQ ID NO: 79









ERTTSIATTT











SEQ ID NO: 80









RTTSIATTTT











SEQ ID NO: 81









TTSIATTTTT











SEQ ID NO: 82









TSIATTTTTT











SEQ ID NO: 83









SIATTTTTTT











SEQ ID NO: 84









IATTTTTTTE











SEQ ID NO: 85









ATTTTTTTES











SEQ ID NO: 86









TTTTTTTESV











SEQ ID NO: 87









TTTTTTESVE











SEQ ID NO: 88









TTTTTESVEE











SEQ ID NO: 89









TTTTESVEEV











SEQ ID NO: 90









TTTESVEEVV











SEQ ID NO: 91









TTESVEEVVR






In some embodiments, the peptide comprises an amino acid sequence selected from 11 consecutive residues of SEQ ID NO: 1, or from the group consisting of the below:









SEQ ID NO: 92









AEESDNVDSAD











SEQ ID NO: 93









EESDNVDSADA











SEQ ID NO: 94









ESDNVDSADAE











SEQ ID NO: 95









SDNVDSADAEE











SEQ ID NO: 96









DNVDSADAEED











SEQ ID NO: 97









NVDSADAEEDD











SEQ ID NO: 98









VDSADAEEDDS











SEQ ID NO: 99









DSADAEEDDSD











SEQ ID NO: 100









SADAEEDDSDV











SEQ ID NO: 101









ADAEEDDSDVW











SEQ ID NO: 102









DAEEDDSDVWW











SEQ ID NO: 103









AEEDDSDVWWG











SEQ ID NO: 104









EEDDSDVWWGG











SEQ ID NO: 105









EDDSDVWWGGA











SEQ ID NO: 106









DDSDVWWGGAD











SEQ ID NO: 107









DSDVWWGGADT











SEQ ID NO: 108









SDVWWGGADTD











SEQ ID NO: 109









DVWWGGADTDY











SEQ ID NO: 110









VWWGGADTDYA











SEQ ID NO: 111









WWGGADTDYAD











SEQ ID NO: 112









WGGADTDYADG











SEQ ID NO: 113









GGADTDYADGS











SEQ ID NO: 114









GADTDYADGSE











SEQ ID NO: 115









ADTDYADGSED











SEQ ID NO: 116









DTDYADGSEDK











SEQ ID NO: 117









TDYADGSEDKV











SEQ ID NO: 118









DYADGSEDKVV











SEQ ID NO: 119









YADGSEDKVVE











SEQ ID NO: 120









ADGSEDKVVEV











SEQ ID NO: 121









DGSEDKVVEVA











SEQ ID NO: 122









GSEDKVVEVAE











SEQ ID NO: 123









SEDKVVEVAEE











SEQ ID NO: 124









EDKVVEVAEEE











SEQ ID NO: 125









DKVVEVAEEEE











SEQ ID NO: 126









KVVEVAEEEEV











SEQ ID NO: 127









VVEVAEEEEVA











SEQ ID NO: 128









VEVAEEEEVAE











SEQ ID NO: 129









EVAEEEEVAEV











SEQ ID NO: 130









VAEEEEVAEVE











SEQ ID NO: 131









AEEEEVAEVEE











SEQ ID NO: 132









EEEEVAEVEEE











SEQ ID NO: 133









EEEVAEVEEEE











SEQ ID NO: 134









EEVAEVEEEEA











SEQ ID NO: 135









EVAEVEEEEAD











SEQ ID NO: 136









VAEVEEEEADD











SEQ ID NO: 137









AEVEEEEADDD











SEQ ID NO: 138









EVEEEEADDDE











SEQ ID NO: 139









VEEEEADDDED











SEQ ID NO: 140









EEEEADDDEDD











SEQ ID NO: 141









EEEADDDEDDE











SEQ ID NO: 142









EEADDDEDDED











SEQ ID NO: 143









EADDDEDDEDG











SEQ ID NO: 144









ADDDEDDEDGD











SEQ ID NO: 145









DDDEDDEDGDE











SEQ ID NO: 146









DDEDDEDGDEV











SEQ ID NO: 147









DEDDEDGDEVE











SEQ ID NO: 148









EDDEDGDEVEE











SEQ ID NO: 149









DDEDGDEVEEE











SEQ ID NO: 150









DEDGDEVEEEA











SEQ ID NO: 151









EDGDEVEEEAE











SEQ ID NO: 152









DGDEVEEEAEE











SEQ ID NO: 153









GDEVEEEAEEP











SEQ ID NO: 154









DEVEEEAEEPY











SEQ ID NO: 155









EVEEEAEEPYE











SEQ ID NO: 156









VEEEAEEPYEE











SEQ ID NO: 157









EEEAEEPYEEA











SEQ ID NO: 158









EEAEEPYEEAT











SEQ ID NO: 159









EAEEPYEEATE











SEQ ID NO: 160









AEEPYEEATER











SEQ ID NO: 161









EEPYEEATERT











SEQ ID NO: 162









EPYEEATERTT











SEQ ID NO: 163









PYEEATERTTS











SEQ ID NO: 164









YEEATERTTSI











SEQ ID NO: 165









EEATERTTSIA











SEQ ID NO: 166









EATERTTSIAT











SEQ ID NO: 167









ATERTTSIATT











SEQ ID NO: 168









TERTTSIATTT











SEQ ID NO: 169









ERTTSIATTTT











SEQ ID NO: 170









RTTSIATTTTT











SEQ ID NO: 171









TTSIATTTTTT











SEQ ID NO: 172









TSIATTTTTTT











SEQ ID NO: 173









SIATTTTTTTE











SEQ ID NO: 174









IATTTTTTTES











SEQ ID NO: 175









ATTTTTTTESV











SEQ ID NO: 176









TTTTTTTESVE











SEQ ID NO: 177









TTTTTTESVEE











SEQ ID NO: 178









TTTTTESVEEV











SEQ ID NO: 179









TTTTESVEEVV











SEQ ID NO: 180









TTTESVEEVVR






In some embodiments, the peptide comprises an amino acid sequence selected from 12 consecutive residues of SEQ ID NO: 1, or from the group consisting of the below:









SEQ ID NO: 181









AEESDNVDSADA











SEQ ID NO: 182









EESDNVDSADAE











SEQ ID NO: 183









ESDNVDSADAEE











SEQ ID NO: 184









SDNVDSADAEED











SEQ ID NO: 185









DNVDSADAEEDD











SEQ ID NO: 186









NVDSADAEEDDS











SEQ ID NO: 187









VDSADAEEDDSD











SEQ ID NO: 188









DSADAEEDDSDV











SEQ ID NO: 189









SADAEEDDSDVW











SEQ ID NO: 190









ADAEEDDSDVWW











SEQ ID NO: 191









DAEEDDSDVWWG











SEQ ID NO: 192









AEEDDSDVWWGG











SEQ ID NO: 193









EEDDSDVWWGGA











SEQ ID NO: 194









EDDSDVWWGGAD











SEQ ID NO: 195









DDSDVWWGGADT











SEQ ID NO: 196









DSDVWWGGADTD











SEQ ID NO: 197









SDVWWGGADTDY











SEQ ID NO: 198









DVWWGGADTDYA











SEQ ID NO: 199









VWWGGADTDYAD











SEQ ID NO: 200









WWGGADTDYADG











SEQ ID NO: 201









WGGADTDYADGS











SEQ ID NO: 202









GGADTDYADGSE











SEQ ID NO: 203









GADTDYADGSED











SEQ ID NO: 204









ADTDYADGSEDK











SEQ ID NO: 205









DTDYADGSEDKV











SEQ ID NO: 206









TDYADGSEDKVV











SEQ ID NO: 207









DYADGSEDKVVE











SEQ ID NO: 208









YADGSEDKVVEV











SEQ ID NO: 209









ADGSEDKVVEVA











SEQ ID NO: 210









DGSEDKVVEVAE











SEQ ID NO: 211









GSEDKVVEVAEE











SEQ ID NO: 212









SEDKVVEVAEEE











SEQ ID NO: 213









EDKVVEVAEEEE











SEQ ID NO: 214









DKVVEVAEEEEV











SEQ ID NO: 215









KVVEVAEEEEVA











SEQ ID NO: 216









VVEVAEEEEVAE











SEQ ID NO: 217









VEVAEEEEVAEV











SEQ ID NO: 218









EVAEEEEVAEVE











SEQ ID NO: 219









VAEEEEVAEVEE











SEQ ID NO: 220









AEEEEVAEVEEE











SEQ ID NO: 221









EEEEVAEVEEEE











SEQ ID NO: 222









EEEVAEVEEEEA











SEQ ID NO: 223









EEVAEVEEEEAD











SEQ ID NO: 224









EVAEVEEEEADD











SEQ ID NO: 225









VAEVEEEEADDD











SEQ ID NO: 226









AEVEEEEADDDE











SEQ ID NO: 227









EVEEEEADDDED











SEQ ID NO: 228









VEEEEADDDEDD











SEQ ID NO: 229









EEEEADDDEDDE











SEQ ID NO: 230









EEEADDDEDDED











SEQ ID NO: 231









EEADDDEDDEDG











SEQ ID NO: 232









EADDDEDDEDGD











SEQ ID NO: 233









ADDDEDDEDGDE











SEQ ID NO: 234









DDDEDDEDGDEV











SEQ ID NO: 235









DDEDDEDGDEVE











SEQ ID NO: 236









DEDDEDGDEVEE











SEQ ID NO: 237









EDDEDGDEVEEE











SEQ ID NO: 238









DDEDGDEVEEEA











SEQ ID NO: 239









DEDGDEVEEEAE











SEQ ID NO: 240









EDGDEVEEEAEE











SEQ ID NO: 241









DGDEVEEEAEEP











SEQ ID NO: 242









GDEVEEEAEEPY











SEQ ID NO: 243









DEVEEEAEEPYE











SEQ ID NO: 244









EVEEEAEEPYEE











SEQ ID NO: 245









VEEEAEEPYEEA











SEQ ID NO: 246









EEEAEEPYEEAT











SEQ ID NO: 247









EEAEEPYEEATE











SEQ ID NO: 248









EAEEPYEEATER











SEQ ID NO: 249









AEEPYEEATERT











SEQ ID NO: 250









EEPYEEATERTT











SEQ ID NO: 251









EPYEEATERTTS











SEQ ID NO: 252









PYEEATERTTSI











SEQ ID NO: 253









YEEATERTTSIA











SEQ ID NO: 254









EEATERTTSIAT











SEQ ID NO: 255









EATERTTSIATT











SEQ ID NO: 256









ATERTTSIATTT











SEQ ID NO: 257









TERTTSIATTTT











SEQ ID NO: 258









ERTTSIATTTTT











SEQ ID NO: 259









RTTSIATTTTTT











SEQ ID NO: 260









TTSIATTTTTTT











SEQ ID NO: 261









TSIATTTTTTTE











SEQ ID NO: 262









SIATTTTTTTES











SEQ ID NO: 263









IATTTTTTTESV











SEQ ID NO: 264









ATTTTTTTESVE











SEQ ID NO: 265









TTTTTTTESVEE











SEQ ID NO: 266









TTTTTTESVEEV











SEQ ID NO: 267









TTTTTESVEEVV











SEQ ID NO: 268









TTTTESVEEVVR






In some embodiments, the peptide comprises an amino acid sequence selected from 13 consecutive residues of SEQ ID NO: 1, or from the group consisting of the below:









SEQ ID NO: 268









TTTTESVEEVVR











SEQ ID NO: 269









AEESDNVDSADAE











SEQ ID NO: 270









EESDNVDSADAEE











SEQ ID NO: 271









ESDNVDSADAEED











SEQ ID NO: 272









SDNVDSADAEEDD











SEQ ID NO: 273









DNVDSADAEEDDS











SEQ ID NO: 274









NVDSADAEEDDSD











SEQ ID NO: 275









VDSADAEEDDSDV











SEQ ID NO: 276









DSADAEEDDSDVW











SEQ ID NO: 277









SADAEEDDSDVWW











SEQ ID NO: 278









ADAEEDDSDVWWG











SEQ ID NO: 279









DAEEDDSDVWWGG











SEQ ID NO: 280









AEEDDSDVWWGGA











SEQ ID NO: 281









EEDDSDVWWGGAD











SEQ ID NO: 282









EDDSDVWWGGADT











SEQ ID NO: 283









DDSDVWWGGADTD











SEQ ID NO: 284









DSDVWWGGADTDY











SEQ ID NO: 285









SDVWWGGADTDYA











SEQ ID NO: 286









DVWWGGADTDYAD











SEQ ID NO: 287









VWWGGADTDYADG











SEQ ID NO: 288









WWGGADTDYADGS











SEQ ID NO: 289









WGGADTDYADGSE











SEQ ID NO: 290









GGADTDYADGSED











SEQ ID NO: 291









GADTDYADGSEDK











SEQ ID NO: 292









ADTDYADGSEDKV











SEQ ID NO: 293









DTDYADGSEDKVV











SEQ ID NO: 294









TDYADGSEDKVVE











SEQ ID NO: 295









DYADGSEDKVVEV











SEQ ID NO: 296









YADGSEDKVVEVA











SEQ ID NO: 297









ADGSEDKVVEVAE











SEQ ID NO: 298









DGSEDKVVEVAEE











SEQ ID NO: 299









GSEDKVVEVAEEE











SEQ ID NO: 300









SEDKVVEVAEEEE











SEQ ID NO: 301









EDKVVEVAEEEEV











SEQ ID NO: 302









DKVVEVAEEEEVA











SEQ ID NO: 303









KVVEVAEEEEVAE











SEQ ID NO: 304









VVEVAEEEEVAEV











SEQ ID NO: 305









VEVAEEEEVAEVE











SEQ ID NO: 306









EVAEEEEVAEVEE











SEQ ID NO: 307









VAEEEEVAEVEEE











SEQ ID NO: 308









AEEEEVAEVEEEE











SEQ ID NO: 309









EEEEVAEVEEEEA











SEQ ID NO: 310









EEEVAEVEEEEAD











SEQ ID NO: 311









EEVAEVEEEEADD











SEQ ID NO: 312









EVAEVEEEEADDD











SEQ ID NO: 313









VAEVEEEEADDDE











SEQ ID NO: 314









AEVEEEEADDDED











SEQ ID NO: 315









EVEEEEADDDEDD











SEQ ID NO: 316









VEEEEADDDEDDE











SEQ ID NO: 317









EEEEADDDEDDED











SEQ ID NO: 318









EEEADDDEDDEDG











SEQ ID NO: 319









EEADDDEDDEDGD











SEQ ID NO: 320









EADDDEDDEDGDE











SEQ ID NO: 321









ADDDEDDEDGDEV











SEQ ID NO: 322









DDDEDDEDGDEVE











SEQ ID NO: 323









DDEDDEDGDEVEE











SEQ ID NO: 324









DEDDEDGDEVEEE











SEQ ID NO: 325









EDDEDGDEVEEEA











SEQ ID NO: 326









DDEDGDEVEEEAE











SEQ ID NO: 327









DEDGDEVEEEAEE











SEQ ID NO: 328









EDGDEVEEEAEEP











SEQ ID NO: 329









DGDEVEEEAEEPY











SEQ ID NO: 330









GDEVEEEAEEPYE











SEQ ID NO: 331









DEVEEEAEEPYEE











SEQ ID NO: 332









EVEEEAEEPYEEA











SEQ ID NO: 333









VEEEAEEPYEEAT











SEQ ID NO: 334









EEEAEEPYEEATE











SEQ ID NO: 335









EEAEEPYEEATER











SEQ ID NO: 336









EAEEPYEEATERT











SEQ ID NO: 337









AEEPYEEATERTT











SEQ ID NO: 338









EEPYEEATERTTS











SEQ ID NO: 339









EPYEEATERTTSI











SEQ ID NO: 340









PYEEATERTTSIA











SEQ ID NO: 341









YEEATERTTSIAT











SEQ ID NO: 342









EEATERTTSIATT











SEQ ID NO: 343









EATERTTSIATTT











SEQ ID NO: 344









ATERTTSIATTTT











SEQ ID NO: 345









TERTTSIATTTTT











SEQ ID NO: 346









ERTTSIATTTTTT











SEQ ID NO: 347









RTTSIATTTTTTT











SEQ ID NO: 348









TTSIATTTTTTTE











SEQ ID NO: 349









TSIATTTTTTTES











SEQ ID NO: 350









SIATTTTTTTESV











SEQ ID NO: 351









IATTTTTTTESVE











SEQ ID NO: 352









ATTTTTTTESVEE











SEQ ID NO: 353









TTTTTTTESVEEV











SEQ ID NO: 354









TTTTTTESVEEVV











SEQ ID NO: 355









TTTTTESVEEVVR






In some embodiments, the peptide comprises an amino acid sequence selected from 14 consecutive residues of SEQ ID NO: 1, or from the group consisting of the below:









SEQ ID NO: 356









AEESDNVDSADAEE











SEQ ID NO: 357









EESDNVDSADAEED











SEQ ID NO: 358









ESDNVDSADAEEDD











SEQ ID NO: 359









SDNVDSADAEEDDS











SEQ ID NO: 360









DNVDSADAEEDDSD











SEQ ID NO: 361









NVDSADAEEDDSDV











SEQ ID NO: 362









VDSADAEEDDSDVW











SEQ ID NO: 363









DSADAEEDDSDVWW











SEQ ID NO: 364









SADAEEDDSDVWWG











SEQ ID NO: 365









ADAEEDDSDVWWGG











SEQ ID NO: 366









DAEEDDSDVWWGGA











SEQ ID NO: 367









AEEDDSDVWWGGAD











SEQ ID NO: 368









EEDDSDVWWGGADT











SEQ ID NO: 369









EDDSDVWWGGADTD











SEQ ID NO: 370









DDSDVWWGGADTDY











SEQ ID NO: 371









DSDVWWGGADTDYA











SEQ ID NO: 372









SDVWWGGADTDYAD











SEQ ID NO: 373









DVWWGGADTDYADG











SEQ ID NO: 374









VWWGGADTDYADGS











SEQ ID NO: 375









WWGGADTDYADGSE











SEQ ID NO: 376









WGGADTDYADGSED











SEQ ID NO: 377









GGADTDYADGSEDK











SEQ ID NO: 378









GADTDYADGSEDKV











SEQ ID NO: 379









ADTDYADGSEDKVV











SEQ ID NO: 380









DTDYADGSEDKVVE











SEQ ID NO: 381









TDYADGSEDKVVEV











SEQ ID NO: 382









DYADGSEDKVVEVA











SEQ ID NO: 383









YADGSEDKVVEVAE











SEQ ID NO: 384









ADGSEDKVVEVAEE











SEQ ID NO: 385









DGSEDKVVEVAEEE











SEQ ID NO: 386









GSEDKVVEVAEEEE











SEQ ID NO: 387









SEDKVVEVAEEEEV











SEQ ID NO: 388









EDKVVEVAEEEEVA











SEQ ID NO: 389









DKVVEVAEEEEVAE











SEQ ID NO: 390









KVVEVAEEEEVAEV











SEQ ID NO: 391









VVEVAEEEEVAEVE











SEQ ID NO: 392









VEVAEEEEVAEVEE











SEQ ID NO: 393









EVAEEEEVAEVEEE











SEQ ID NO: 394









VAEEEEVAEVEEEE











SEQ ID NO: 395









AEEEEVAEVEEEEA











SEQ ID NO: 396









EEEEVAEVEEEEAD











SEQ ID NO: 397









EEEVAEVEEEEADD











SEQ ID NO: 398









EEVAEVEEEEADDD











SEQ ID NO: 399









EVAEVEEEEADDDE











SEQ ID NO: 400









VAEVEEEEADDDED











SEQ ID NO: 401









AEVEEEEADDDEDD











SEQ ID NO: 402









EVEEEEADDDEDDE











SEQ ID NO: 403









VEEEEADDDEDDED











SEQ ID NO: 404









EEEEADDDEDDEDG











SEQ ID NO: 405









EEEADDDEDDEDGD











SEQ ID NO: 406









EEADDDEDDEDGDE











SEQ ID NO: 407









EADDDEDDEDGDEV











SEQ ID NO: 408









ADDDEDDEDGDEVE











SEQ ID NO: 409









DDDEDDEDGDEVEE











SEQ ID NO: 410









DDEDDEDGDEVEEE











SEQ ID NO: 411









DEDDEDGDEVEEEA











SEQ ID NO: 412









EDDEDGDEVEEEAE











SEQ ID NO: 413









DDEDGDEVEEEAEE











SEQ ID NO: 414









DEDGDEVEEEAEEP











SEQ ID NO: 415









EDGDEVEEEAEEPY











SEQ ID NO: 416









DGDEVEEEAEEPYE











SEQ ID NO: 417









GDEVEEEAEEPYEE











SEQ ID NO: 418









DEVEEEAEEPYEEA











SEQ ID NO: 419









EVEEEAEEPYEEAT











SEQ ID NO: 420









VEEEAEEPYEEATE











SEQ ID NO: 421









EEEAEEPYEEATER











SEQ ID NO: 422









EEAEEPYEEATERT











SEQ ID NO: 423









EAEEPYEEATERTT











SEQ ID NO: 424









AEEPYEEATERTTS











SEQ ID NO: 425









EEPYEEATERTTSI











SEQ ID NO: 426









EPYEEATERTTSIA











SEQ ID NO: 427









PYEEATERTTSIAT











SEQ ID NO: 428









YEEATERTTSIATT











SEQ ID NO: 429









EEATERTTSIATTT











SEQ ID NO: 430









EATERTTSIATTTT











SEQ ID NO: 431









ATERTTSIATTTTT











SEQ ID NO: 432









TERTTSIATTTTTT











SEQ ID NO: 433









ERTTSIATTTTTTT











SEQ ID NO: 434









RTTSIATTTTTTTE











SEQ ID NO: 435









TTSIATTTTTTTES











SEQ ID NO: 436









TSIATTTTTTTESV











SEQ ID NO: 437









SIATTTTTTTESVE











SEQ ID NO: 438









IATTTTTTTESVEE











SEQ ID NO: 439









ATTTTTTTESVEEV











SEQ ID NO: 440









TTTTTTTESVEEVV











SEQ ID NO: 441









TTTTTTESVEEVVR






In some embodiments, the peptide comprises an amino acid sequence selected from 24 consecutive residues of SEQ ID NO: 1, or from the group consisting of the below:









SEQ ID NO: 900









ATERTTSIATTTTTTTESVEEVVR






In some embodiments, the peptide is a fragment of the APP-binding domain of PTPσ. Therefore, in some embodiments, the peptide is a fragment of SEQ ID NO:442, as listed below, which has at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or more amino acids, or a conservative variant thereof. The underlined amino acids represent residues in the ligand-binding pocket.









(SEQ ID NO: 442)







EEPPRFIKEPKDQIGVSGGVASFVCQATGDPKPRVTWNKKGKKVNSQRFE





TIEFDESAGAVLRIQPLRTPRDENVYECVAQNSVGEITVHAKLTVLRE.






Therefore, in some embodiments, the peptide comprises an amino acid sequence selected from 10 consecutive residues of SEQ ID NO: 442, or from the group consisting of the below:









SEQ ID NO: 443









EEPPRFIKEP











SEQ ID NO: 444









EPPRFIKEPK











SEQ ID NO: 445









PPRFIKEPKD











SEQ ID NO: 446









PRFIKEPKDQ











SEQ ID NO: 447









RFIKEPKDQI











SEQ ID NO: 448









FIKEPKDQIG











SEQ ID NO: 449









IKEPKDQIGV











SEQ ID NO: 450









KEPKDQIGVS











SEQ ID NO: 451









EPKDQIGVSG











SEQ ID NO: 452









PKDQIGVSGG











SEQ ID NO: 453









KDQIGVSGGV











SEQ ID NO: 454









DQIGVSGGVA











SEQ ID NO: 455









QIGVSGGVAS











SEQ ID NO: 456









IGVSGGVASF











SEQ ID NO: 457









GVSGGVASFV











SEQ ID NO: 458









VSGGVASFVC











SEQ ID NO: 459









SGGVASFVCQ











SEQ ID NO: 460









GGVASFVCQA











SEQ ID NO: 461









GVASFVCQAT











SEQ ID NO: 462









VASFVCQATG











SEQ ID NO: 463









ASFVCQATGD











SEQ ID NO: 464









SFVCQATGDP











SEQ ID NO: 465









FVCQATGDPK











SEQ ID NO: 466









VCQATGDPKP











SEQ ID NO: 467









CQATGDPKPR











SEQ ID NO: 468









QATGDPKPRV











SEQ ID NO: 469









ATGDPKPRVT











SEQ ID NO: 470









TGDPKPRVTW











SEQ ID NO: 471









GDPKPRVTWN











SEQ ID NO: 472









DPKPRVTWNK











SEQ ID NO: 473









PKPRVTWNKK











SEQ ID NO: 474









KPRVTWNKKG











SEQ ID NO: 475









PRVTWNKKGK











SEQ ID NO: 476









RVTWNKKGKK











SEQ ID NO: 477









VTWNKKGKKV











SEQ ID NO: 478









TWNKKGKKVN











SEQ ID NO: 479









WNKKGKKVNS











SEQ ID NO: 480









NKKGKKVNSQ











SEQ ID NO: 481









KKGKKVNSQR











SEQ ID NO: 482









KGKKVNSQRF











SEQ ID NO: 483









GKKVNSQRFE











SEQ ID NO: 484









KKVNSQRFET











SEQ ID NO: 485









KVNSQRFETI











SEQ ID NO: 486









VNSQRFETIE











SEQ ID NO: 487









NSQRFETIEF











SEQ ID NO: 488









SQRFETIEFD











SEQ ID NO: 489









QRFETIEFDE











SEQ ID NO: 490









RFETIEFDES











SEQ ID NO: 491









FETIEFDESA











SEQ ID NO: 492









ETIEFDESAG











SEQ ID NO: 493









TIEFDESAGA











SEQ ID NO: 494









IEFDESAGAV











SEQ ID NO: 495









EFDESAGAVL











SEQ ID NO: 496









FDESAGAVLR











SEQ ID NO: 497









DESAGAVLRI











SEQ ID NO: 498









ESAGAVLRIQ











SEQ ID NO: 499









SAGAVLRIQP











SEQ ID NO: 500









AGAVLRIQPL











SEQ ID NO: 501









GAVLRIQPLR











SEQ ID NO: 502









AVLRIQPLRT











SEQ ID NO: 503









VLRIQPLRTP











SEQ ID NO: 504









LRIQPLRTPR











SEQ ID NO: 505









RIQPLRTPRD











SEQ ID NO: 506









IQPLRTPRDE











SEQ ID NO: 507









QPLRTPRDEN











SEQ ID NO: 508









PLRTPRDENV











SEQ ID NO: 509









LRTPRDENVY











SEQ ID NO: 510









RTPRDENVYE











SEQ ID NO: 511









TPRDENVYEC











SEQ ID NO: 512









PRDENVYECV











SEQ ID NO: 513









RDENVYECVA











SEQ ID NO: 514









DENVYECVAQ











SEQ ID NO: 515









ENVYECVAQN











SEQ ID NO: 516









NVYECVAQNS











SEQ ID NO: 517









VYECVAQNSV











SEQ ID NO: 518









YECVAQNSVG











SEQ ID NO: 519









ECVAQNSVGE











SEQ ID NO: 520









CVAQNSVGEI











SEQ ID NO: 521









VAQNSVGEIT











SEQ ID NO: 522









AQNSVGEITV











SEQ ID NO: 523









QNSVGEITVH











SEQ ID NO: 524









NSVGEITVHA











SEQ ID NO: 525









SVGEITVHAK











SEQ ID NO: 526









VGEITVHAKL











SEQ ID NO: 527









GEITVHAKLT











SEQ ID NO: 528









EITVHAKLTV











SEQ ID NO: 529









ITVHAKLTVL











SEQ ID NO: 530









TVHAKLTVLR











SEQ ID NO: 531









VHAKLTVLRE






In some embodiments, the peptide comprises an amino acid sequence selected from 11 consecutive residues of SEQ ID NO: 442, or from the group consisting of the below:









SEQ ID NO: 531









VHAKLTVLRE











SEQ ID NO: 532









EEPPRFIKEPK











SEQ ID NO: 533









EPPRFIKEPKD











SEQ ID NO: 534









PPRFIKEPKDQ











SEQ ID NO: 535









PRFIKEPKDQI











SEQ ID NO: 536









RFIKEPKDQIG











SEQ ID NO: 537









FIKEPKDQIGV











SEQ ID NO: 538









IKEPKDQIGVS











SEQ ID NO: 539









KEPKDQIGVSG











SEQ ID NO: 540









EPKDQIGVSGG











SEQ ID NO: 541









PKDQIGVSGGV











SEQ ID NO: 542









KDQIGVSGGVA











SEQ ID NO: 543









DQIGVSGGVAS











SEQ ID NO: 544









QIGVSGGVASF











SEQ ID NO: 545









IGVSGGVASFV











SEQ ID NO: 546









GVSGGVASFVC











SEQ ID NO: 547









VSGGVASFVCQ











SEQ ID NO: 548









SGGVASFVCQA











SEQ ID NO: 549









GGVASFVCQAT











SEQ ID NO: 550









GVASFVCQATG











SEQ ID NO: 551









VASFVCQATGD











SEQ ID NO: 552









ASFVCQATGDP











SEQ ID NO: 553









SFVCQATGDPK











SEQ ID NO: 554









FVCQATGDPKP











SEQ ID NO: 555









VCQATGDPKPR











SEQ ID NO: 556









CQATGDPKPRV











SEQ ID NO: 557









QATGDPKPRVT











SEQ ID NO: 558









ATGDPKPRVTW











SEQ ID NO: 559









TGDPKPRVTWN











SEQ ID NO: 560









GDPKPRVTWNK











SEQ ID NO: 561









DPKPRVTWNKK











SEQ ID NO: 562









PKPRVTWNKKG











SEQ ID NO: 563









KPRVTWNKKGK











SEQ ID NO: 564









PRVTWNKKGKK











SEQ ID NO: 565









RVTWNKKGKKV











SEQ ID NO: 566









VTWNKKGKKVN











SEQ ID NO: 567









TWNKKGKKVNS











SEQ ID NO: 568









WNKKGKKVNSQ











SEQ ID NO: 569









NKKGKKVNSQR











SEQ ID NO: 570









KKGKKVNSQRF











SEQ ID NO: 571









KGKKVNSQRFE











SEQ ID NO: 572









GKKVNSQRFET











SEQ ID NO: 573









KKVNSQRFETI











SEQ ID NO: 574









KVNSQRFETIE











SEQ ID NO: 575









VNSQRFETIEF











SEQ ID NO: 576









NSQRFETIEFD











SEQ ID NO: 577









SQRFETIEFDE











SEQ ID NO: 578









QRFETIEFDES











SEQ ID NO: 579









RFETIEFDESA











SEQ ID NO: 580









FETTEFDESAG











SEQ ID NO: 581









ETIEFDESAGA











SEQ ID NO: 582









TIEFDESAGAV











SEQ ID NO: 583









IEFDESAGAVL











SEQ ID NO: 584









EFDESAGAVLR











SEQ ID NO: 585









FDESAGAVLRI











SEQ ID NO: 586









DESAGAVLRIQ











SEQ ID NO: 587









ESAGAVLRIQP











SEQ ID NO: 588









SAGAVLRIQPL











SEQ ID NO: 589









AGAVLRIQPLR











SEQ ID NO: 590









GAVLRIQPLRT











SEQ ID NO: 591









AVLRIQPLRTP











SEQ ID NO: 592









VLRIQPLRTPR











SEQ ID NO: 593









LRIQPLRTPRD











SEQ ID NO: 594









RIQPLRTPRDE











SEQ ID NO: 595









IQPLRTPRDEN











SEQ ID NO: 596









QPLRTPRDENV











SEQ ID NO: 597









PLRTPRDENVY











SEQ ID NO: 598









LRTPRDENVYE











SEQ ID NO: 599









RTPRDENVYEC











SEQ ID NO: 600









TPRDENVYECV











SEQ ID NO: 601









PRDENVYECVA











SEQ ID NO: 602









RDENVYECVAQ











SEQ ID NO: 603









DENVYECVAQN











SEQ ID NO: 604









ENVYECVAQNS











SEQ ID NO: 605









NVYECVAQNSV











SEQ ID NO: 606









VYECVAQNSVG











SEQ ID NO: 607









YECVAQNSVGE











SEQ ID NO: 608









ECVAQNSVGEI











SEQ ID NO: 609









CVAQNSVGEIT











SEQ ID NO: 610









VAQNSVGEITV











SEQ ID NO: 611









AQNSVGEITVH











SEQ ID NO: 612









QNSVGEITVHA











SEQ ID NO: 613









NSVGEITVHAK











SEQ ID NO: 614









SVGEITVHAKL











SEQ ID NO: 615









VGEITVHAKLT











SEQ ID NO: 616









GEITVHAKLTV











SEQ ID NO: 617









EITVHAKLTVL











SEQ ID NO: 618









ITVHAKLTVLR











SEQ ID NO: 619









TVHAKLTVLRE






In some embodiments, the peptide comprises an amino acid sequence selected from 12 consecutive residues of SEQ ID NO: 442, or from the group consisting of the below:









SEQ ID NO: 620









EEPPRFIKEPKD











SEQ ID NO: 621









EPPRFIKEPKDQ











SEQ ID NO: 622









PPRFIKEPKDQI











SEQ ID NO: 623









PRFIKEPKDQIG











SEQ ID NO: 624









RFIKEPKDQIGV











SEQ ID NO: 625









FIKEPKDQIGVS











SEQ ID NO: 626









IKEPKDQIGVSG











SEQ ID NO: 627









KEPKDQIGVSGG











SEQ ID NO: 628









EPKDQIGVSGGV











SEQ ID NO: 629









PKDQIGVSGGVA











SEQ ID NO: 630









KDQIGVSGGVAS











SEQ ID NO: 631









DQIGVSGGVASF











SEQ ID NO: 632









QIGVSGGVASFV











SEQ ID NO: 633









IGVSGGVASFVC











SEQ ID NO: 634









GVSGGVASFVCQ











SEQ ID NO: 635









VSGGVASFVCQA











SEQ ID NO: 636









SGGVASFVCQAT











SEQ ID NO: 637









GGVASFVCQATG











SEQ ID NO: 638









GVASFVCQATGD











SEQ ID NO: 639









VASFVCQATGDP











SEQ ID NO: 640









ASFVCQATGDPK











SEQ ID NO: 641









SFVCQATGDPKP











SEQ ID NO: 642









FVCQATGDPKPR











SEQ ID NO: 643









VCQATGDPKPRV











SEQ ID NO: 644









CQATGDPKPRVT











SEQ ID NO: 645









QATGDPKPRVTW











SEQ ID NO: 646









ATGDPKPRVTWN











SEQ ID NO: 647









TGDPKPRVTWNK











SEQ ID NO: 648









GDPKPRVTWNKK











SEQ ID NO: 649









DPKPRVTWNKKG











SEQ ID NO: 650









PKPRVTWNKKGK











SEQ ID NO: 651









KPRVTWNKKGKK











SEQ ID NO: 652









PRVTWNKKGKKV











SEQ ID NO: 653









RVTWNKKGKKVN











SEQ ID NO: 654









VTWNKKGKKVNS











SEQ ID NO: 655









TWNKKGKKVNSQ











SEQ ID NO: 656









WNKKGKKVNSQR











SEQ ID NO: 657









NKKGKKVNSQRF











SEQ ID NO: 658









KKGKKVNSQRFE











SEQ ID NO: 659









KGKKVNSQRFET











SEQ ID NO: 660









GKKVNSQRFETI











SEQ ID NO: 661









KKVNSQRFETIE











SEQ ID NO: 662









KVNSQRFETIEF











SEQ ID NO: 663









VNSQRFETIEFD











SEQ ID NO: 664









NSQRFETIEFDE











SEQ ID NO: 665









SQRFETIEFDES











SEQ ID NO: 666









QRFETIEFDESA











SEQ ID NO: 667









RFETIEFDESAG











SEQ ID NO: 668









FETIEFDESAGA











SEQ ID NO: 669









ETIEFDESAGAV











SEQ ID NO: 670









TIEFDESAGAVL











SEQ ID NO: 671









IEFDESAGAVLR











SEQ ID NO: 672









EFDESAGAVLRI











SEQ ID NO: 673









FDESAGAVLRIQ











SEQ ID NO: 674









DESAGAVLRIQP











SEQ ID NO: 675









ESAGAVLRIQPL











SEQ ID NO: 676









SAGAVLRIQPLR











SEQ ID NO: 677









AGAVLRIQPLRT











SEQ ID NO: 678









GAVLRIQPLRTP











SEQ ID NO: 679









AVLRIQPLRTPR











SEQ ID NO: 680









VLRIQPLRTPRD











SEQ ID NO: 681









LRIQPLRTPRDE











SEQ ID NO: 682









RIQPLRTPRDEN











SEQ ID NO: 683









IQPLRTPRDENV











SEQ ID NO: 684









QPLRTPRDENVY











SEQ ID NO: 685









PLRTPRDENVYE











SEQ ID NO: 686









LRTPRDENVYEC











SEQ ID NO: 687









RTPRDENVYECV











SEQ ID NO: 688









TPRDENVYECVA











SEQ ID NO: 689









PRDENVYECVAQ











SEQ ID NO: 690









RDENVYECVAQN











SEQ ID NO: 691









DENVYECVAQNS











SEQ ID NO: 692









ENVYECVAQNSV











SEQ ID NO: 693









NVYECVAQNSVG











SEQ ID NO: 694









VYECVAQNSVGE











SEQ ID NO: 695









YECVAQNSVGEI











SEQ ID NO: 696









ECVAQNSVGEIT











SEQ ID NO: 697









CVAQNSVGEITV











SEQ ID NO: 698









VAQNSVGEITVH











SEQ ID NO: 699









AQNSVGEITVHA











SEQ ID NO: 700









QNSVGEITVHAK











SEQ ID NO: 701









NSVGEITVHAKL











SEQ ID NO: 702









SVGEITVHAKLT











SEQ ID NO: 703









VGEITVHAKLTV











SEQ ID NO: 704









GEITVHAKLTVL











SEQ ID NO: 705









EITVHAKLTVLR











SEQ ID NO: 706









ITVHAKLTVLRE






In some embodiments, the peptide comprises an amino acid sequence selected from 13 consecutive residues of SEQ ID NO: 442, or from the group consisting of the below:









SEQ ID NO: 707









EEPPRFIKEPKDQ











SEQ ID NO: 708









EPPRFIKEPKDQI











SEQ ID NO: 709









PPRFIKEPKDQIG











SEQ ID NO: 710









PRFIKEPKDQIGV











SEQ ID NO: 711









RFIKEPKDQIGVS











SEQ ID NO: 712









FIKEPKDQIGVSG











SEQ ID NO: 713









IKEPKDQIGVSGG











SEQ ID NO: 714









KEPKDQIGVSGGV











SEQ ID NO: 715









EPKDQIGVSGGVA











SEQ ID NO: 716









PKDQIGVSGGVAS











SEQ ID NO: 717









KDQIGVSGGVASF











SEQ ID NO: 718









DQIGVSGGVASFV











SEQ ID NO: 719









QIGVSGGVASFVC











SEQ ID NO: 720









IGVSGGVASFVCQ











SEQ ID NO: 721









GVSGGVASFVCQA











SEQ ID NO: 722









VSGGVASFVCQAT











SEQ ID NO: 723









SGGVASFVCQATG











SEQ ID NO: 724









GGVASFVCQATGD











SEQ ID NO: 725









GVASFVCQATGDP











SEQ ID NO: 726









VASFVCQATGDPK











SEQ ID NO: 727









ASFVCQATGDPKP











SEQ ID NO: 728









SFVCQATGDPKPR











SEQ ID NO: 729









FVCQATGDPKPRV











SEQ ID NO: 730









VCQATGDPKPRVT











SEQ ID NO: 731









CQATGDPKPRVTW











SEQ ID NO: 732









QATGDPKPRVTWN











SEQ ID NO: 733









ATGDPKPRVTWNK











SEQ ID NO: 734









TGDPKPRVTWNKK











SEQ ID NO: 735









GDPKPRVTWNKKG











SEQ ID NO: 736









DPKPRVTWNKKGK











SEQ ID NO: 737









PKPRVTWNKKGKK











SEQ ID NO: 738









KPRVTWNKKGKKV











SEQ ID NO: 739









PRVTWNKKGKKVN











SEQ ID NO: 740









RVTWNKKGKKVNS











SEQ ID NO: 741









VTWNKKGKKVNSQ











SEQ ID NO: 742









TWNKKGKKVNSQR











SEQ ID NO: 743









WNKKGKKVNSQRF











SEQ ID NO: 744









NKKGKKVNSQRFE











SEQ ID NO: 745









KGKKVNSQRFET











SEQ ID NO: 746









KGKKVNSQRFETI











SEQ ID NO: 747









GKKVNSQRFETIE











SEQ ID NO: 748









KKVNSQRFETIEF











SEQ ID NO: 749









KVNSQRFETIEFD











SEQ ID NO: 750









VNSQRFETIEFDE











SEQ ID NO: 751









NSQRFETIEFDES











SEQ ID NO: 752









SQRFETIEFDESA











SEQ ID NO: 753









QRFETIEFDESAG











SEQ ID NO: 754









RFETIEFDESAGA











SEQ ID NO: 755









FETIEFDESAGAV











SEQ ID NO: 756









ETIEFDESAGAVL











SEQ ID NO: 757









TIEFDESAGAVLR











SEQ ID NO: 758









IEFDESAGAVLRI











SEQ ID NO: 759









EFDESAGAVLRIQ











SEQ ID NO: 760









FDESAGAVLRIQP











SEQ ID NO: 761









DESAGAVLRIQPL











SEQ ID NO: 762









ESAGAVLRIQPLR











SEQ ID NO: 763









SAGAVLRIQPLRT











SEQ ID NO: 764









AGAVLRIQPLRTP











SEQ ID NO: 765









GAVLRIQPLRTPR











SEQ ID NO: 766









AVLRIQPLRTPRD











SEQ ID NO: 767









VLRIQPLRTPRDE











SEQ ID NO: 768









LRIQPLRTPRDEN











SEQ ID NO: 769









RIQPLRTPRDENV











SEQ ID NO: 770









IQPLRTPRDENVY











SEQ ID NO: 771









QPLRTPRDENVYE











SEQ ID NO: 772









PLRTPRDENVYEC











SEQ ID NO: 773









LRTPRDENVYECV











SEQ ID NO: 774









RTPRDENVYECVA











SEQ ID NO: 775









TPRDENVYECVAQ











SEQ ID NO: 776









PRDENVYECVAQN











SEQ ID NO: 777









RDENVYECVAQNS











SEQ ID NO: 778









DENVYECVAQNSV











SEQ ID NO: 779









ENVYECVAQNSVG











SEQ ID NO: 780









NVYECVAQNSVGE











SEQ ID NO: 781









VYECVAQNSVGEI











SEQ ID NO: 782









YECVAQNSVGEIT











SEQ ID NO: 783









ECVAQNSVGEITV











SEQ ID NO: 784









CVAQNSVGEITVH











SEQ ID NO: 785









VAQNSVGEITVHA











SEQ ID NO: 786









AQNSVGEITVHAK











SEQ ID NO: 787









QNSVGEITVHAKL











SEQ ID NO: 788









NSVGEITVHAKLT











SEQ ID NO: 789









SVGEITVHAKLTV











SEQ ID NO: 790









VGEITVHAKLTVL











SEQ ID NO: 791









GEITVHAKLTVLR











SEQ ID NO: 792









EITVHAKLTVLRE






In some embodiments, the peptide comprises an amino acid sequence selected from 14 consecutive residues of SEQ ID NO: 442, or from the group consisting of the below:









SEQ ID NO: 793









EEPPRFIKEPKDQI











SEQ ID NO: 794









EPPRFIKEPKDQIG











SEQ ID NO: 795









PPRFIKEPKDQIGV











SEQ ID NO: 796









PRFIKEPKDQIGVS











SEQ ID NO: 797









RFIKEPKDQIGVSG











SEQ ID NO: 798









FIKEPKDQIGVSGG











SEQ ID NO: 799









IKEPKDQIGVSGGV











SEQ ID NO: 800









KEPKDQIGVSGGVA











SEQ ID NO: 801









EPKDQIGVSGGVAS











SEQ ID NO: 802









PKDQIGVSGGVASF











SEQ ID NO: 803









KDQIGVSGGVASFV











SEQ ID NO: 804









DQIGVSGGVASFVC











SEQ ID NO: 805









QIGVSGGVASFVCQ











SEQ ID NO: 806









IGVSGGVASFVCQA











SEQ ID NO: 807









GVSGGVASFVCQAT











SEQ ID NO: 808









VSGGVASFVCQATG











SEQ ID NO: 809









SGGVASFVCQATGD











SEQ ID NO: 810









GGVASFVCQATGDP











SEQ ID NO: 811









GVASFVCQATGDPK











SEQ ID NO: 812









VASFVCQATGDPKP











SEQ ID NO: 813









ASFVCQATGDPKPR











SEQ ID NO: 814









SFVCQATGDPKPRV











SEQ ID NO: 815









FVCQATGDPKPRVT











SEQ ID NO: 816









VCQATGDPKPRVTW











SEQ ID NO: 817









CQATGDPKPRVTWN











SEQ ID NO: 818









QATGDPKPRVTWNK











SEQ ID NO: 819









ATGDPKPRVTWNKK











SEQ ID NO: 820









TGDPKPRVTWNKKG











SEQ ID NO: 821









GDPKPRVTWNKKGK











SEQ ID NO: 822









DPKPRVTWNKKGKK











SEQ ID NO: 823









PKPRVTWNKKGKKV











SEQ ID NO: 824









KPRVTWNKKGKKVN











SEQ ID NO: 825









PRVTWNKKGKKVNS











SEQ ID NO: 826









RVTWNKKGKKVNSQ











SEQ ID NO: 827









VTWNKKGKKVNSQR











SEQ ID NO: 828









TWNKKGKKVNSQRF











SEQ ID NO: 829









WNKKGKKVNSQRFE











SEQ ID NO: 830









NKKGKKVNSQRFET











SEQ ID NO: 831









KKGKKVNSQRFETI











SEQ ID NO: 832









KGKKVNSQRFETIE











SEQ ID NO: 833









GKKVNSQRFETIEF











SEQ ID NO: 834









KKVNSQRFETIEFD











SEQ ID NO: 835









KVNSQRFETIEFDE











SEQ ID NO: 836









VNSQRFETIEFDES











SEQ ID NO: 837









NSQRFETIEFDESA











SEQ ID NO: 838









SQRFETIEFDESAG











SEQ ID NO: 839









QRFETIEFDESAGA











SEQ ID NO: 840









RFETIEFDESAGAV











SEQ ID NO: 841









FETIEFDESAGAVL











SEQ ID NO: 842









ETIEFDESAGAVLR











SEQ ID NO: 843









TIEFDESAGAVLRI











SEQ ID NO: 844









IEFDESAGAVLRIQ











SEQ ID NO: 845









EFDESAGAVLRIQP











SEQ ID NO: 846









FDESAGAVLRIQPL











SEQ ID NO: 847









DESAGAVLRIQPLR











SEQ ID NO: 848









ESAGAVLRIQPLRT











SEQ ID NO: 849









SAGAVLRIQPLRTP











SEQ ID NO: 850









AGAVLRIQPLRTPR











SEQ ID NO: 851









GAVLRIQPLRTPRD











SEQ ID NO: 852









AVLRIQPLRTPRDE











SEQ ID NO: 853









VLRIQPLRTPRDEN











SEQ ID NO: 854









LRIQPLRTPRDENV











SEQ ID NO: 855









RIQPLRTPRDENVY











SEQ ID NO: 856









IQPLRTPRDENVYE











SEQ ID NO: 857









QPLRTPRDENVYEC











SEQ ID NO: 858









PLRTPRDENVYECV











SEQ ID NO: 859









LRTPRDENVYECVA











SEQ ID NO: 860









RTPRDENVYECVAQ











SEQ ID NO: 861









TPRDENVYECVAQN











SEQ ID NO: 862









PRDENVYECVAQNS











SEQ ID NO: 863









RDENVYECVAQNSV











SEQ ID NO: 864









DENVYECVAQNSVG











SEQ ID NO: 865









ENVYECVAQNSVGE











SEQ ID NO: 866









NVYECVAQNSVGEI











SEQ ID NO: 867









VYECVAQNSVGEIT











SEQ ID NO: 868









YECVAQNSVGEITV











SEQ ID NO: 869









ECVAQNSVGEITVH











SEQ ID NO: 870









CVAQNSVGEITVHA











SEQ ID NO: 871









VAQNSVGEITVHAK











SEQ ID NO: 872









AQNSVGEITVHAKL











SEQ ID NO: 873









QNSVGEITVHAKLT











SEQ ID NO: 874









NSVGEITVHAKLTV











SEQ ID NO: 875









SVGEITVHAKLTVL











SEQ ID NO: 876









VGEITVHAKLTVLR











SEQ ID NO: 877









GEITVHAKLTVLRE






In some embodiments, the disclosed peptide further comprises a blood brain barrier penetrating sequence. For example, cell-penetrating peptides (CPPs) are a group of peptides, which have the ability to cross cell membrane bilayers. CPPs themselves can exert biological activity and can be formed endogenously. Fragmentary studies demonstrate their ability to enhance transport of different cargoes across the blood-brain barrier (BBB). The cellular internalization sequence can be any cell-penetrating peptide sequence capable of penetrating the BBB. Non-limiting examples of CPPs include Polyarginine (e.g., R9), Antennapedia sequences, TAT, HIV-Tat, Penetratin, Antp-3A (Antp mutant), Buforin II, Transportan, MAP (model amphipathic peptide), K-FGF, Ku70, Prion, pVEC, Pep-1, SynB1, Pep-7, HN-1, BGSC (Bis-Guanidinium-Spermidine-Cholesterol, and BGTC (Bis-Guanidinium-Tren-Cholesterol) (see Table 1).









TABLE 1







Cell Internalization Transporters









Name
Sequence
SEQ ID NO





Polyarginine
RRRRRRRRR
SEQ ID NO: 878





Antp
RQPKIWFPNRRKPWKK
SEQ ID NO: 879





HIV-Tat
GRKKRRQRPPQ
SEQ ID NO: 880





Penetratin
RQIKIWFQNRRMKWKK
SEQ ID NO: 881





Antp-3A
RQIAIWFQNRRMKWAA
SEQ ID NO: 882





Tat
RKKRRQRRR
SEQ ID NO: 883





Buforin II
TRSSRAGLQFPVGRVHRLLRK
SEQ ID NO: 884





Transportan
GWTLNSAGYLLGKINKALAALA
SEQ ID NO: 885



KKIL





model
KLALKLALKALKAALKLA
SEQ ID NO: 886


amphipathic


peptide (MAP)





K-FGF
AAVALLPAVLLALLAP
SEQ ID NO: 887





Ku70
VPMLK-PMLKE
SEQ ID NO: 888





Prion
MANLGYWLLALFVTMWTDVGLC
SEQ ID NO: 889



KKRPKP





pVEC
LLIILRRRIRKQAHAHSK
SEQ ID NO: 890





Pep-1
KETWWETWWTEWSQPKKKRKV
SEQ ID NO: 891





SynB1
RGGRLSYSRRRFSTSTGR
SEQ ID NO: 892





Pep-7
SDLWEMMMVSLACQY
SEQ ID NO: 893





HN-1
TSPLNIHNGQKL
SEQ ID NO: 894





Tat
GRKKRRQRRRPQ
SEQ ID NO: 895





Tat
RKKRRQRRRC
SEQ ID NO: 896









Therefore, in some embodiments, the disclosed peptide is a fusion protein, e.g., containing the APP-binding domain of PTPσ, the PTPσ-binding domain of APP, or a combination thereof and a CPP. Fusion proteins, also known as chimeric proteins, are proteins created through the joining of two or more genes, which originally coded for separate proteins. Translation of this fusion gene results in a single polypeptide with function properties derived from each of the original proteins. Recombinant fusion proteins can be created artificially by recombinant DNA technology for use in biological research or therapeutics.


In some embodiments, linker (or “spacer”) peptides are also added which make it more likely that the proteins fold independently and behave as expected. Linkers in protein or peptide fusions are sometimes engineered with cleavage sites for proteases or chemical agents which enable the liberation of the two separate proteins. This technique is often used for identification and purification of proteins, by fusing a GST protein, FLAG peptide, or a hexa-his peptide (aka: a 6×his-tag) which can be isolated using nickel or cobalt resins (affinity chromatography). Chimeric proteins can also be manufactured with toxins or antibodies attached to them in order to study disease development.


Compositions that Restore Molecular Balance of CS and HS in the Perineuronal Space:


Chondroitin sulfates (CS) and heparin or its analog heparan sulfates (HS) are two main classes of glycosaminoglycans (GAGs) in the brain that are sensed by neurons via Receptor Protein Tyrosine8. The ratio of CS and HS therefore affects the downstream effects of PTPσ, because CS and HS compete to interact with the receptor yet lead to opposite signaling and neuronal responses (such as neurite regeneration). CS increases but HS decreases APP β-cleavage products (Example 2). Therefore, methods involving administering to the subject a composition that restore the physiological molecular CS/HS balance may be used to treat and prevent aforementioned neurodegenerative diseases. These therapies could be applied alternatively or in addition to the polypeptides listed above. In some embodiments, administering HS, or its analog heparin, or their mimetics modified to reduce anti-coagulant effect, with a saccharide chain length of 17, 18, 19, 20, 21, 22, 23, 24 units or longer, could assist in restoring the physiological molecular CS/HS balance. In some embodiments, the balance is restored by administering enzymes that digest CS (such as ChABC) or prevent the degradation of HS (such as Heparanase inhibitors PI-88, OGT 2115, or PG545). Alternatively or in addition, agents that mimic the HS/heparin effect of PTPσ clustering8, such as multivalent antibodies, could be administered.


Pharmaceutical Compositions


The peptides disclosed can be used therapeutically in combination with a pharmaceutically acceptable carrier. Pharmaceutical carriers suitable for administration of the compounds provided herein include any such carriers known to those skilled in the art to be suitable for the particular mode of administration. The carrier would naturally be selected to minimize any degradation of the active ingredient and to minimize any adverse side effects in the subject, as would be well known to one of skill in the art.


In some embodiments, the peptides described above are formulated into pharmaceutical compositions using techniques and procedures well known in the art (See, e.g., Ansel, Introduction to Pharmaceutical Dosage Forms, 4th Edition, 1985, 126).


Liquid pharmaceutically administrable compositions can, for example, be prepared by dissolving, dispersing, or otherwise mixing an active compound as defined above and optional pharmaceutical adjuvants in a carrier, such as, for example, water, saline, aqueous dextrose, glycerol, glycols, ethanol, and the like, to thereby form a solution or suspension.


Dosage forms or compositions containing active ingredient in the range of 0.005% to 100% with the balance made up from non-toxic carrier may be prepared. Methods for preparation of these compositions are known to those skilled in the art. The contemplated compositions may contain 0.001%-100% active ingredient, or in one embodiment 0.1-95%.


Methods of Screening


Also disclosed are methods of screening for candidate compounds that slow, stop, reverse, or prevent neurodegeneration.


Methods of Screening Based on APP-PTPσ Binding:

In some embodiments, the method comprising providing a sample comprising APP and PTPσ in an environment permissive for APP-PTPσ binding, contacting the sample with a candidate compound, and assaying the sample for APP-PTPσ binding, wherein a decrease in APP-PTPσ binding compared to control values is an indication that the candidate agent is effective to slow, stop, reverse, or prevent neurodegeneration.


The binding of PTPσ to APP can be detected using routine methods that do not disturb protein binding.


In some embodiments, the binding of PTPσ to APP can be detected using immunodetection methods. The steps of various useful immunodetection methods have been described in the scientific literature, such as, e.g., Maggio et al., Enzyme-Immunoassay, (1987) and Nakamura, et al., Enzyme Immunoassays: Heterogeneous and Homogeneous Systems, Handbook of Experimental Immunology, Vol. 1: Immunochemistry, 27.1-27.20 (1986), each of which is incorporated herein by reference in its entirety and specifically for its teaching regarding immunodetection methods. Immunoassays, in their most simple and direct sense, are binding assays involving binding between antibodies and antigen. Examples of immunoassays are enzyme linked immunosorbent assays (ELISAs), radioimmunoassays (RIA), radioimmune precipitation assays (RIPA), immunobead capture assays, Western blotting, dot blotting, gel-shift assays, Flow cytometry, protein arrays, multiplexed bead arrays, magnetic capture, in vivo imaging, fluorescence resonance energy transfer (FRET), and fluorescence recovery/localization after photobleaching (FRAP/FLAP).


The methods can be cell-based or cell-free assays.


In some embodiments, the binding between PTPσ and APP can be detected using fluorescence activated cell sorting (FACS). For example, disclosed are cell lines transfected with of PTPσ and APP fused to fluorescent proteins. These cell lines can facilitate high-throughput screens for biologically expressed and chemically synthesized molecules that disrupt the binding between PTPσ and APP.


In some embodiments, the binding between PTPσ and APP can be detected in a cell-free setting where one of these two binding partners is purified and immobilized/captured through covalent or non-covalent bond to a solid surface or beads, while the other binding partner is allowed to bind in the presence of biologically expressed and chemically synthesized molecules to screen candidate agents for their efficacies in dissociating APP-PTPσ interaction.


In some embodiments, the binding between PTPσ and APP can be detected in a setting where cell membrane preparations extracted from fresh rodent brain homogenates (containing both APP and PTPσ) are contacted with biologically expressed and chemically synthesized molecules. Subsequently, one of the binding partners is immunoprecipitated and the binding or co-immunoprecipitation of the other binding partner is detected using its specific antibody.


A candidate agent that decreases or abolishes APP-PTPσ binding in a disclosed method herein has the potential to slow, stop, reverse, or prevent neurodegeneration.


Methods of Screening Based on APP Amyloidogenic Processing:

In some embodiments, the method comprising contacting/incubating a candidate compound with cell membrane preparations extracted from fresh rodent brain homogenates, wherein a decrease in APP β- and/or γ-cleavage products is an indication that the candidate agent has the potential to slow, stop, reverse, or prevent neurodegeneration. APP β- and/or γ-cleavage products can be detected by routine biochemical methods such as Western blot analysis, ELISA, and immnuopurification.


Libraries of Molecules and Compounds:

In general, candidate agents can be identified from large libraries of natural products or synthetic (or semi-synthetic) extracts or chemical libraries according to methods known in the art. Those skilled in the field of drug discovery and development will understand that the precise source of test extracts or compounds is not critical to the screening procedure(s) used.


Accordingly, virtually any number of chemical extracts or compounds can be screened using the exemplary methods described herein. Examples of such extracts or compounds include, but are not limited to, plant-, fungal-, prokaryotic- or animal-based extracts, fermentation broths, and synthetic compounds, as well as modification of existing compounds. Numerous methods are also available for generating random or directed synthesis (e.g., semi-synthesis or total synthesis) of any number of chemical compounds, including, but not limited to, saccharide-, lipid-, peptide-, and nucleic acid-based compounds. Synthetic compound libraries are commercially available, e.g., from purveyors of chemical libraries including but not limited to ChemBridge Corporation (16981 Via Tazon, Suite G, San Diego, Calif., 92127, USA, www.chembridge.com); ChemDiv (6605 Nancy Ridge Drive, San Diego, Calif. 92121, USA); Life Chemicals (1103 Orange Center Road, Orange, Conn. 06477); Maybridge (Trevillett, Tintagel, Cornwall PL34 OHW, UK).


Alternatively, libraries of natural compounds in the form of bacterial, fungal, plant, and animal extracts are commercially available from a number of sources, including 02H, (Cambridge, UK), MerLion Pharmaceuticals Pte Ltd (Singapore Science Park II, Singapore 117528) and Galapagos NV (Generaal De Wittelaan L11 A3, B-2800 Mechelen, Belgium).


In addition, natural and synthetically produced libraries are produced, if desired, according to methods known in the art, e.g., by standard extraction and fractionation methods or by standard synthetic methods in combination with solid phase organic synthesis, micro-wave synthesis and other rapid throughput methods known in the art to be amenable to making large numbers of compounds for screening purposes. Furthermore, if desired, any library or compound, including sample format and dissolution is readily modified and adjusted using standard chemical, physical, or biochemical methods.


Candidate agents encompass numerous chemical classes, but are most often organic molecules, e.g., small organic compounds having a molecular weight of more than 100 and less than about 2,500 Daltons, or, in some embodiments, having a molecular weight of more than 100 and less than about 5,000 Daltons. Candidate agents can include functional groups necessary for structural interaction with proteins, particularly hydrogen bonding, and typically include at least an amine, carbonyl, hydroxyl or carboxyl group, for example, at least two of the functional chemical groups. The candidate agents often contain cyclical carbon or heterocyclic structures and/or aromatic or polyaromatic structures substituted with one or more of the above functional groups.


In some embodiments, the candidate agents are proteins. In some aspects, the candidate agents are naturally occurring proteins or fragments of naturally occurring proteins. Thus, for example, cellular extracts containing proteins, or random or directed digests of proteinaceous cellular extracts, can be used. In this way libraries of procaryotic and eucaryotic proteins can be made for screening using the methods herein. The libraries can be bacterial, fungal, viral, and vertebrate proteins, and human proteins.


Methods of Treatment


Disclosed herein are methods for treating neurodegenerative diseases that involve β-amyloid pathologies and/or Tau pathologies, including but not limited to Alzheimer's disease, Lewy body dementia, frontotemporal dementia, cerebral amyloid angiopathy, primary age-related tauopathy, chronic traumatic encephalopathy, Parkinson's disease, postencephalitic parkinsonism, Huntington's disease, amyolateral sclerosis, Pick's disease, progressive supranuclear palsy, corticobasal degeneration, Lytico-Bodig disease, ganglioglioma and gangliocytoma, subacute sclerosing panencephalitis, Hallervorden-Spatz disease, and/or Creutzfeldt-Jakob disease.


These peptides, compositions, and methods may also be used to prevent these neurodegenerative diseases in populations at risk, such as people with Down syndrome and those suffered from brain injuries or cerebral ischemia, as well as the aging population.


In some embodiments, these methods involve disrupting the binding between PTPσ and APP, preventing β-amyloidogenic processing of APP without affecting other major substrates of β- and γ-secretases. For example, the methods can involve administering to a subject a peptide disclosed herein. In other embodiments, monoclonal antibodies could be formed against the IG1 domain of PTPσ or a fragment thereof a fragment between the E1 and E2 domain of the APP695 isoform, or both, and these antibodies, or fragments thereof, could be administered to the subject.


Chondroitin sulfates (CS) and heparin or its analog heparan sulfates (HS) are two main classes of glycosaminoglycans (GAGs) in the brain that are “sensed” by neurons via Receptor Protein Tyrosine8. The ratio of CS and HS therefore affects the downstream effects of PTPσ, because CS and HS compete to interact with the receptor yet lead to opposite signaling and neuronal responses (such as neurite regeneration). CS increases but HS decreases APP β-cleavage products (Example 2). Therefore, in some embodiments, the methods involve administering to the subject a composition, which restores the physiological molecular CS/HS balance, may be used to treat and prevent aforementioned neurodegenerative diseases. These therapies could be applied alternatively or in addition to the polypeptides listed above. In some embodiments, administering HS, or its analog heparin, or their mimetics modified to reduce anti-coagulant effects, with a saccharide chain length of 17, 18, 19, 20, 21, 22, 23, 24 units or longer, could assist in restoring the physiological molecular CS/HS balance. In some embodiments, the balance is restored by administering enzymes that digest CS (such as Chondroitinase ABC) or prevent the degradation of HS (such as Heparanase inhibitors PI-88, OGT 2115, or PG545). Alternatively or in addition, agents that mimic the HS/heparin effect of PTPσ clustering8, such as multivalent antibodies, could be administered.


In some embodiments, the method involves administering a composition described herein in a dose equivalent to parenteral administration of about 0.1 ng to about 100 g per kg of body weight, about 10 ng to about 50 g per kg of body weight, about 100 ng to about 1 g per kg of body weight, from about 1 μg to about 100 mg per kg of body weight, from about 1 μg to about 50 mg per kg of body weight, from about 1 mg to about 500 mg per kg of body weight; and from about 1 mg to about 50 mg per kg of body weight. Alternatively, the amount of composition administered to achieve a therapeutic effective dose is about 0.1 ng, 1 ng, 10 ng, 100 ng, 1 μg, 10 μg 100 μg, 1 mg, 2 mg, 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 mg, 10 mg, 11 mg, 12 mg, 13 mg, 14 mg, 15 mg, 16 mg, 17 mg, 18 mg, 19 mg, 20 mg, 30 mg, 40 mg, 50 mg, 60 mg, 70 mg, 80 mg, 90 mg, 100 mg, 500 mg per kg of body weight or greater.


A number of embodiments of the invention have been described. Nevertheless, it will be understood that various modifications may be made without departing from the spirit and scope of the invention. Accordingly, other embodiments are within the scope of the following claims.


EXAMPLES
Example 1: Alzheimer's Disease Pathogenesis is Dependent on Neuronal Receptor PTPσ

Methods and Materials


Mouse Lines:


Mice were maintained under standard conditions approved by the Institutional Animal Care and Use Committee. Wild type and PTPσ-deficient mice of Balb/c background were provided by Dr. Michel L. Tremblay9. Homozygous TgAPP-SwDI mice, C57BL/6-Tg(Thy1-APPSwDutIowa)BWevn/Mmjax, stock number 007027, were from the Jackson Laboratory. These mice express human APP transgene harboring Swedish, Dutch, and Iowa mutations, and were bred with Balb/c mice heterozygous for the PTPσ gene to generate bigenic mice heterozygous for both TgAPP-SwDI and PTPσ genes, which are hybrids of 50% C57BL/6J and 50% Balb/c genetic background. These mice were further bred with Balb/c mice heterozygous for the PTPσ gene. The offspring from this mating are used in experiments, which include littermates of the following genotypes: TgAPP-SwDI(+/−)PTPσ(+/+), mice heterozygous for TgAPP-SwDI transgene with wild type PTPσ; TgAPP-SwDI(+/−)PTPσ(−/−), mice heterozygous for TgAPP-SwDI transgene with genetic depletion of PTPσ; TgAPP-SwDI(−/−) PTPσ(+/+), mice free of TgAPP-SwDI transgene with wild type PTPσ. Both TgAPP-SwDI(−/−) PTPσ(+/+) and Balb/c PTPσ(+/+) are wild type mice but with different genetic background. Heterozygous TgAPP-SwInd (J20) mice, 6.Cg-Tg(PDGFB-APPSwInd)20Lms/2Mmjax, were provided by Dr. Lennart Mucke. These mice express human APP transgene harboring Swedish and Indiana mutations, and were bred with the same strategy as described above to obtain mice with genotypes of TgAPP-SwInd (+/−)PTPσ(+/+) and TgAPP-SwInd (+/−)PTPσ(−/−).


Antibodies:


















Application
Clone #
Catalog #
Supplier




















Primary Antibodies






Mouse anti-Actin
WB
AC-40
A4700
Sigma-Aldrich


Rabbit anti-APH1
WB

PA5-20318
Thermo Scientific


Rabbit anti-APP C-term
WB, IP, IHC
Y188
NB110-55461
Novus Biologicals


Mouse anti-murine Aβ, 1-16
WB, IP
M3.2
805701
Biolegend


Mouse anti-human Aβ, 1-16
WB, IP, IHC, ELISA
6E10
803001
Biolegend


Mosue anti-Aβ, 17-24
WB, IHC
4G8
SIG-39220
Biolegend


Mouse HRP-conjugated anti-Aβ 1-40
ELISA
11A50-B10
SIG-39146
Biolegend


Mouse HRP-conjugated anti-Aβ 1-42
ELISA
12F4
805507
Biolegend


Rabbit anti-BACE1 C-Term, B690
WB

PRB-617C
Covance


Guinea Pig anti-BACE1 C-Term
IP

840201
Biolegend


Chiken anti-GFAP
IHC

ab4674
Abcam


Rabbit anti-Neuregulin
WB

sc-348
Santa Cruz Biotechnology


Rabbit anti-Nicastrin
WB

5665
Cell Signaling


Rabbit anti-Notch NICD (val1 744)
WB

4147
Cell Signaling


Rabbit anti-Notch (C-20)
WB

sc-6014R
Santa Cruz Biotechnology


Rabbit anti-PEN2
WB

8598
Cell Signaling


Rabbit anti-Presenilin 1/2 NTF
WB

840201
Abcam


Rabbit anti-Presenilin 1 CTF
WB

5643
Cell Signaling


Rabbit anti-Presenilin 2 CTF
WB

9979
Cell Signaling


Mouse anti-PTPσ ICD
WB, IHC
17G7.2
MM-002-P
Medimabs


Mouse anti-PTPσ ECD
WB

ab55640
Abcam


Rabbit anti-Synaptophysin
IHC

AB9272
Millipore


Mouse anti-Tau
WB, IHC
Tau-5
MAB361
Millipore


Mouse anti-Tau
IHC
Tau-46
4019
Cell Signaling


Secondary and Tertiary Antibodies


Goat anti-mouse IgG HRP-conjugated
WB

7076S
Cell Signaling


Goat anti-rabbit IgG HRP-conjugated
WB

7074S
Cell Signaling


Goat anti-mouse IgG Alexa488
IHC

A-11001
Invitrogen


Donkey anti-goat IgG Alexa488
IHC

A-11055
Invitrogen


Chicken anti-rabbit IgG CF568
IHC

SAB4600426
Sigma-Aldrich


Donkey anti-chicken IgG Cy3
IHC

703-165-155
JacksonImmunoResearch









Immunohistochemistry:


Adult rat and mice were perfused intracardially with fresh made 4% paraformaldehyde in cold phosphate-buffered saline (PBS). The brains were collected and post-fixed for 2 days at 4° C. Paraffin embedded sections of 10 μM thickness were collected for immunostaining. The sections were deparaffinized and sequentially rehydrated. Antigen retrieval was performed at 100° C. in Tris-EDTA buffer (pH 9.0) for 50 min. Sections were subsequently washed with distilled water and PBS, incubated at room temperature for 1 hour in blocking buffer (PBS, with 5% normal donkey serum, 5% normal goat serum, and 0.2% Triton X-100). Primary antibody incubation was performed in a humidified chamber at 4° C. overnight. After 3 washes in PBS with 0.2% Triton X-100, the sections were then incubated with a mixture of secondary and tertiary antibodies at room temperature for 2 hours. All antibodies were diluted in blocking buffer with concentrations recommended by the manufacturers. Mouse primary antibodies were detected by goat anti-mouse Alexa488 together with donkey anti-goat Alexa488 antibodies; rabbit primary antibodies were detected by chicken anti-rabbit CF568 and donkey anti-chicken Cy3 antibodies; chicken antibody was detected with donkey anti-chicken Cy3 antibody. Sections stained with only secondary and tertiary antibodies (without primary antibodies) were used as negative controls. At last, DAPI (Invitrogen, 300 nM) was applied on sections for nuclear staining. Sections were washed 5 times before mounted in Fluoromount (SouthernBiotech).


Wide field and confocal images were captured using Zeiss Axio Imager M2 and LSM780, respectively. Images are quantified using the Zen 2 Pro software and ImageJ.


Protein Extraction, Immunoprecipitation, and Western Blot Analysis:


For the co-immunoprecipitation of APP and PTPσ, RIPA buffer was used (50 mM Tris-HCl, pH 8.0, 1 mM EDTA, 150 mM NaCl, 1% NP40, 0.1% SDS, 0.5% sodium deoxycholate). For the co-immunoprecipitation of APP and BACE1, NP40 buffer was used (50 mM Tris-HCl, pH 8.0, 1 mM EDTA, 150 mM NaCl, 1% NP40) without or with SDS at concentration of 0.1%, 0.3%, and 0.4%. For total protein extraction and immunopurification of CTFβ, SDS concentration in RIPA buffer was adjusted to 1% to ensure protein extraction from the lipid rafts. Mouse or rat forebrains were homogenized thoroughly on ice in homogenization buffers (as mention above) containing protease and phosphatase inhibitors (Thermo Scientific). For each half of forebrain, buffer volume of at least 5 ml for mouse and 8 ml for rat was used to ensure sufficient detergent/tissue ratio. The homogenates were incubated at 4° C. for 1 hour with gentle mixing, sonicated on ice for 2 minutes in a sonic dismembrator (Fisher Scientific Model 120, with pulses of 50% output, 1 second on and 1 second off), followed with another hour of gentle mixing at 4° C. All samples were used fresh without freezing and thawing.


For co-immunoprecipitation and immunopurification, the homogenates were then centrifuged at 85,000×g for 1 hour at 4° C. and the supernatants were collected. Protein concentration was measured using BCA Protein Assay Kit (Thermo Scientific). 0.5 mg total proteins of brain homogenates were incubated with 5 μg of designated antibody and 30 μl of Protein-A sepharose beads (50% slurry, Roche), in a total volume of 1 ml adjusted with RIPA buffer. Samples were gently mixed at 4° C. overnight. Subsequently, the beads were washed 5 times with cold immunoprecipitation buffer. Samples were then incubated in Laemmli buffer with 100 mM of DTT at 75° C. for 20 minutes and subjected to western blot analysis.


For analysis of protein expression level, the homogenates were centrifuged at 23,000×g for 30 min at 4° C. and the supernatants were collected. Protein concentration was measured using BCA Protein Assay Kit (Thermo Scientific). 30 μg of total proteins were subjected to western blot analysis.


Electrophoresis of protein samples was conducted using 4-12% Bis-Tris Bolt Plus Gels, with either MOPS or MES buffer and Novex Sharp Pre-stained Protein Standard (all from Invitrogen). Proteins were transferred to nitrocellulose membrane (0.2 μm pore size, Bio-Rad) and blotted with selected antibodies (see table above) at concentrations suggested by the manufacturers. Primary antibodies were diluted in SuperBlock TBS Blocking Buffer (Thermo Scientific) and incubated with the nitrocellulose membranes at 4° C. overnight; secondary antibodies were diluted in PBS with 5% nonfat milk and 0.2% Tween20 and incubated at room temperature for 2 hours. Membranes were washes 4 times in PBS with 0.2% Tween20 between primary and secondary antibodies and before chemiluminescent detection with SuperSignal West Pico Chemiluminescent Substrate (Thermo Scientific).


Western blot band intensity was quantified by densitometry.


Aβ ELISA Assays:


Mouse forebrains were thoroughly homogenized in tissue homogenization buffer (2 mM Tris pH 7.4, 250 mM sucrose, 0.5 mM EDTA, 0.5 mM EGTA) containing protease inhibitor cocktail (Roche), followed by centrifugation at 135,000×g (33,500 RPM with SW50.1 rotor) for 1 hour at 4° C. Proteins in the pellets were extracted with formic acid (FA) and centrifuged at 109,000×g (30,100 RPM with SW50.1 rotor) for 1 hour at 4° C. The supernatants were collected and diluted 1:20 in neutralization buffer (1 M Tris base, 0.5 M Na2HPO4, 0.05% NaN3) and subsequently 1:3 in ELISA buffer (PBS with 0.05% Tween-20, 1% BSA, and 1 mM AEBSF). Diluted samples were loaded onto ELISA plates pre-coated with 6E10 antibody (Biolegend) to capture Aβ peptides. Serial dilutions of synthesized human Aβ 1-40 or 1-42 (American Peptide) were loaded to determine a standard curve. Aβ was detected using an HRP labeled antibody for either Aβ 1-40 or 1-42 (see table above). ELISA was developed using TMB substrate (Thermo Scientific) and reaction was stopped with IN HCl. Plates were read at 450 nm and concentrations of Aβ in samples were determined using the standard curve.


Behavior Assays:


The Y-maze assay: Mice were placed in the center of the Y-maze and allowed to move freely through each arm. Their exploratory activities were recorded for 5 minutes. An arm entry is defined as when all four limbs are within the arm. For each mouse, the number of triads is counted as “spontaneous alternation”, which was then divided by the number of total arm entries, yielding a percentage score. The novel object test: On day 1, mice were exposed to empty cages (45 cm×24 cm×22 cm) with blackened walls to allow exploration and habituation to the arena. During day 2 to day 4, mice were returned to the same cage with two identical objects placed at an equal distance. On each day mice were returned to the cage at approximately the same time during the day and allowed to explore for 10 minutes. Cages and objects were cleaned with 70% ethanol between each animal. Subsequently, 2 hours after the familiarization session on day 4, mice were put back to the same cage where one of the familiar objects (randomly chosen) was replaced with a novel object, and allowed to explore for 5 minutes. Mice were scored using Observer software (Noldus) on their time duration and visiting frequency exploring either object. Object exploration was defined as facing the object and actively sniffing or touching the object, whereas any climbing behavior was not scored. The discrimination indexes reflecting interest in the novel object is denoted as either the ratio of novel object exploration to total object exploration (NO/NO+FO) or the ratio of novel object exploration to familiar object exploration (NO/FO). All tests and data analyses were conducted in a double-blinded manner.


Statistics:


2-tailed Student's t test was used for two-group comparison. Relationship between two variables was analyzed using linear regression. All error bars show standard error of the means (SEM).


Results


PTPσ is an APP Binding Partner in the Brain.


Previously identified as a neuronal receptor of extracellular proteoglycans8,10,11, PTPσ is expressed throughout the adult nervous system, most predominantly in the hippocampus12,13, one of earliest affected brain regions in AD. Using immunohistochemistry and confocal imaging, it was found that PTPσ and APP (the precursor of Aβ) colocalize in hippocampal pyramidal neurons of adult rat brains, most intensively in the initial segments of apical dendrites, and in the perinuclear and axonal regions with a punctate pattern (FIG. 1a-f). To assess whether this colocalization reflects a binding interaction between these two molecules, co-immunoprecipitation experiments were run from brain homogenates. In brains of rats and mice with different genetic background, using various antibodies of APP and PTPσ, a fraction of PTPσ that co-immunoprecipitates with APP was consistently detected, providing evidence of a molecular complex between these two transmembrane proteins (FIG. 1h, i; FIG. 2).


Genetic Depletion of PTPσ Reduces β-Amyloidogenic Products of APP.


The molecular interaction between PTPσ and APP prompted an investigation on whether PTPσ plays a role in amyloidogenic processing of APP. In neurons, APP is mainly processed through alternative cleavage by either α- or β-secretase. These secretases release the N-terminal portion of APP from its membrane-tethering C-terminal fragment (CTFα or CTFβ, respectively), which can be further processed by the γ-secretase14,15 Sequential cleavage of APP by the β- and γ-secretases is regarded as amyloidogenic processing since it produces Aβ peptides16. When overproduced, the Aβ peptides can form soluble oligomers that trigger ramification of cytotoxic cascades, whereas progressive aggregation of Aβ eventually results in the formation of senile plaques in the brains of AD patients (FIG. 3a). To test the effect of PTPσ in this amyloidogenic processing the levels of APP β- and γ-cleavage products in mouse brains were analyzed, with or without PTPσ.


Western blot analysis with protein extracts from mouse brains showed that genetic depletion of PTPσ does not affect the expression level of full length APP (FIG. 3b; FIG. 4a). However, an antibody against the C-terminus of APP detects a band at a molecular weight consistent with CTFβ, which is reduced in PTPσ-deficient mice as compared to their age-sex-matched wild type littermates (FIG. 3b). Additionally, in two AD mouse models expressing human APP genes with amyloidogenic mutations17,18, a similar decrease of an APP CTF upon PTPσ depletion was observed (FIG. 3b; FIG. 4b). The TgAPP-SwDI and TgAPP-SwInd mice, each expressing a human APP transgene harboring the Swedish mutation near the β-cleavage site, were crossed with the PTPσ line to generate offsprings that are heterozygous for their respective APP transgene, with or without PTPσ. Because the Swedish mutation carried by these APP transgenes is prone to β-cleavage, the predominant form of APP CTF in these transgenic mice is predicted to be CTFβ. Thus, the reduction of APP CTF in PTPσ-deficient APP transgenic mice may indicate a regulatory role of PTPσ on CTFβ level. However, since the APP C-terminal antibody used in these experiments can recognize both CTFα and CTFβ, as well as the phosphorylated species of these CTFs (longer exposure of western blots showed multiple CTF bands), judging the identity of the reduced CTF simply by its molecular weight may be inadequate. CTFβ immunopurification was therefore performed with subsequent western blot detection, using an antibody that recognizes CTFβ but not CTFα (FIG. 3c, d; FIG. 4c, d). With this method, we confirmed that PTPσ depletion decreases the level of CTFβ originated from both mouse endogenous and human transgenic APP.


Because CTFβ is an intermediate proteolytic product between β- and γ-cleavage, its decreased steady state level could result from either reduced production by β-cleavage or increased degradation by subsequent γ-secretase cleavage (FIG. 3a). To distinguish between these two possibilities, the level of Aβ peptides was measured, because they are downstream products from CTFβ degradation by γ-cleavage. Using ELISA assays with brain homogenates from the TgAPP-SwDI mice, it was found that PTPσ depletion decreases the levels of Aβ peptides to a similar degree as that of CTFβ (FIG. 3e, f). Consistently, as Aβ peptides gradually aggregate into plaques during aging of the transgenic mice, a substantial decrease of cerebral Aβ deposition was observed in APP transgenic PTPσ-deficient mice as compared to the age-matched APP transgenic littermates expressing wild type PTPσ (FIG. 3g, h; FIG. 4e, f). Thus, the concurrent decrease of β- and γ-cleavage products argues against an increased γ-secretase activity, but instead suggests a reduced β-secretase cleavage of APP, which suppresses not only the level of CTFβ but also downstream Aβ production in PTPσ-deficient brains.


Curtailed Progression of β-Amyloidosis in the Absence of PTPσ.


Progressive cerebral Aβ aggregation (β-amyloidosis) is regarded as a benchmark of AD progression. To investigate the effects of PTPσ on this pathological development, Aβ deposits in the brains of 9-month old (mid-aged) and 16-month old (aged) TgAPP-SwDI mice were monitored. At age of 9 to 11 months, Aβ deposits are found predominantly in the hippocampus, especially in the hilus of the dentate gyrus (DG) (FIG. 3g, h). By 16 months, the pathology spreads massively throughout the entire brain. The propagation of Aβ deposition, however, is curbed by genetic depletion of PTPσ, as quantified using the DG hilus as a representative area (FIG. 3i). Between the ages of 9 and 16 months, the Aβ burden is more than doubled in TgAPP-SwDI mice expressing wild type PTPσ [APP-SwDI(+)PTPσ(+/+)], but only shows marginal increase in the transgenic mice lacking functional PTPσ [APP-SwDI(+)PTPσ(−/−)]. Meanwhile, the Aβ loads measured in 9-month old APP-SwDI(+)PTPσ(+/+) mice are similar to those of 16-month old APP-SwDI(+)PTPσ(−/−) mice (p=0.95), indicating a restraint of disease progression by PTPσ depletion (FIG. 3i).


Decreased BACE1-APP Affinity in PTPσ-Deficient Brains.


Consistent with these observations that suggest a facilitating role of PTPσ in APP β-cleavage, the data further reveal that PTPσ depletion weakens the interaction of APP with BACE1, the β-secretase in the brain. To test the in vivo affinity between BACE1 and APP, co-immunoprecipitation were performed of the enzyme and substrate from mouse brain homogenates in buffers with serially increased detergent stringency. Whereas BACE1-APP association is nearly equal in wild type and PTPσ-deficient brains under mild buffer conditions, increasing detergent stringency in the buffer unveils that the molecular complex is more vulnerable to dissociation in brains without PTPσ (FIG. 5). Thus a lower BACE1-APP affinity in PTPσ-deficient brains may likely be an underlying mechanism for the decreased levels of CTFβ and its derivative Aβ.


Although it cannot be ruled out that some alternative uncharacterized pathway may contribute to the parallel decrease of CTFβ and Aβ in PTPσ-deficient brains, these data consistently support the notion that PTPσ regulates APP amyloidogenic processing, likely via facilitation of BACE1 activity on APP, the initial process of Aβ production.


The Specificity of β-Amyloidogenic Regulation by PTPσ.


The constraining effect of PTPσ on APP amyloidogenic products led to further questions regarding whether this observation reflects a specific regulation of APP metabolism, or alternatively, a general modulation on the β- and γ-secretases. First, the expression level of these secretases in mouse brains were assessed with or without PTPσ. No change was found for BACE1 or the essential subunits of γ-secretase (FIG. 6a, b). Additionally, the question of whether PTPσ broadly modulates β- and γ-secretase activities was tested by examining the proteolytic processing of their other substrates. Besides APP, Neuregulinl (NRG1)19-21 and Notch22-24 are the major in vivo substrates of BACE1 and γ-secretase, respectively. Neither BACE1 cleavage of NRG1 nor γ-secretase cleavage of Notch is affected by PTPσ deficiency (FIG. 6c, d). Taken together, these data rule out a generic modulation of β- and γ-secretases, but rather suggest a specificity of APP amyloidogenic regulation by PTPσ.


PTPσ Depletion Relieves Neuroinflammation and Synaptic Impairment in APP Transgenic Mice.


Substantial evidence from earlier studies has established that overproduction of Aβ in the brain elicits multiplex downstream pathological events, including chronic inflammatory responses of the glia, such as persistent astrogliosis. The reactive (inflammatory) glia would then crosstalk with neurons, evoking a vicious feedback loop that amplifies neurodegeneration during disease progression25-27.


The TgAPP-SwDI model is one of the earliest to develop neurodegenerative pathologies and behavioral deficits among many existing AD mouse models17. These mice were therefore chosen to further examine the role of PTPσ in AD pathologies downstream of neurotoxic Aβ.


The APP-SwDI(+)PTPσ(+/+) mice, which express the TgAPP-SwDI transgene and wild type PTPσ, have developed severe neuroinflammation in the brain by the age of 9 months, as measured by the level of GFAP (glial fibrillary acidic protein), a marker of astrogliosis (FIG. 7). In the DG hilus, for example, GFAP expression level in the APP-SwDI(+)PTPσ(+/+) mice is more than tenfold compared to that in age-matched non-transgenic littermates [APP-SwDI(−) PTPσ(+/+)]. PTPσ deficiency, however, effectively attenuates astrogliosis induced by the amyloidogenic transgene. In the APP-SwDI(+)PTPσ(−/−) brains, depletion of PTPσ restores GFAP expression in DG hilus back to a level close to that of non-transgenic wild type littermates (FIG. 7k).


Among all brain regions, the most affected by the expression of TgAPP-SwDI transgene appears to be the hilus of the DG, where Aβ deposition and astrogliosis are both found to be the most severe (FIG. 3g, h; FIG. 7). The question was therefore raised whether the pathologies in this area have an impact on the mossy fiber axons of DG pyramidal neurons, which project through the hilus into the CM region, where they synapse with the CM dendrites. Upon examining the presynaptic markers in CA3 mossy fiber terminal zone, decreased levels of Synaptophysin and Synapsin-1 were found in the APP-SwDI(+)PTPσ(+/+) mice, comparing to their age-matched non-transgenic littermates (FIG. 8, data not shown for Synapsin-1). Such synaptic impairment, evidently resulting from the expression of the APP transgene and possibly the overproduction of Aβ, is reversed by genetic depletion of PTPσ in the APP-SwDI(+)PTPσ(−/−) mice (FIG. 8).


Interestingly, the APP-SwDI(+)PTPσ(−/−) mice sometimes express higher levels of presynaptic markers in the CA3 terminal zone than their age-matched non-transgenic wild type littermates (FIG. 8g). This observation, although not statistically significant, may suggest an additional synaptic effect of PTPσ that is independent of the APP transgene, as observed in previous studies28.


Tau Pathology in Aging AD Mouse Brains is Dependent on PTPσ.


Neurofibrillary tangles composed of hyperphosphorylated and aggregated Tau are commonly found in AD brains. These tangles tend to develop in a hierarchical pattern, appearing first in the entorhinal cortex before spreading to other brain regions5,6. The precise mechanism of tangle formation, however, is poorly understood. The fact that Tau tangles and Aβ deposits can be found in separate locations in postmortem brains has led to the question of whether Tau pathology in AD is independent of Aβ accumulation5,6. Additionally, despite severe cerebral β-amyloidosis in many APP transgenic mouse models, Tau tangles have not been reported, further questioning the relationship between Aβ and Tau pathologies in vivo.


Nonetheless, a few studies did show non-tangle like assemblies of Tau in dystrophic neurites surrounding Aβ plaques in APP transgenic mouse lines29-31, arguing that Aβ can be a causal factor for Tau dysregulation, despite that the precise nature of Tau pathologies may be different between human and mouse. In the histological analysis using an antibody against the proline-rich domain of Tau, Tau aggregation was observed in the brains of both TgAPP-SwDI and TgAPP-SwInd mice during the course of aging (around 9 months for the APP-SwDI(+)PTPσ(+/+) mice and 15 months for the APP-SwInd(+)PTPσ(+/+) mice) (FIG. 9; FIG. 10). Such aggregation is not seen in aged-matched non-transgenic littermates (FIG. 9h), suggesting that it is a pathological event downstream from the expression of amyloidogenic APP transgenes, possibly a result of Aβ cytotoxicity. Genetic depletion of PTPσ, which diminishes Aβ levels, suppresses Tau aggregation in both TgAPP-SwDI and TgAPP-SwInd mice (FIG. 9; FIG. 10).


In both TgAPP-SwDI and TgAPP-SwInd mice, the Tau aggregates are found predominantly in the molecular layer of the piriform and entorhinal cortices, and occasionally in the hippocampal region (FIG. 9; FIG. 10), reminiscent of the early stage tangle locations in AD brains32. Upon closer examination, the Tau aggregates are often found in punctate shapes, likely in debris from degenerated cell bodies and neurites, scattered in areas free of nuclear staining (FIG. 11a-f). Rarely, a few are in fibrillary structures, probably in degenerated cells before disassembling (FIG. 11g, h). To confirm these findings, an additional antibody was used to recognize the C-terminus of Tau. The same morphologies (FIG. 11i) and distribution pattern (FIG. 9a) were detected.


Consistent with the findings in postmortem AD brains, the distribution pattern of Tau aggregates in the TgAPP-SwDI brain does not correlate with that of Aβ deposition, which is pronounced in the hippocampus yet only sporadic in the piriform or entorhinal cortex at the age of 9 months (FIG. 3g, h). Given that the causation of Tau pathology in these mice is possibly related to the overproduced Aβ, the segregation of predominant areas for Aβ and Tau depositions may indicate that the cytotoxicity originates from soluble Aβ instead of the deposited amyloid. It is also evident that neurons in different brain regions are not equally vulnerable to developing Tau pathology.


Next, the question of whether the expression of APP transgenes or genetic depletion of PTPσ regulates Tau aggregation by changing its expression level and/or phosphorylation status was examined. Western blot analysis of brain homogenates showed that Tau protein expression is not affected by the APP transgenes or PTPσ (FIG. 12), suggesting that the aggregation may result from local misfolding of Tau rather than an overexpression of this protein. These experiments with brain homogenates also revealed that TgAPP-SwDI or TgAPP-SwInd transgene, which apparently causes Tau aggregation, does not enhance the phosphorylation of Tau residues including Serine191, Therionine194, and Therionine220 (data not shown), whose homologues in human Tau (Serine202, Therionine205, and Therionine231) are typically hyperphosphorylated in neurofibrillary tangles. These findings are consistent with a recent quantitative study showing similar post-translational modifications of Tau in wild type and TgAPP-SwInd mice33. Furthermore, unlike previously reported29,30, we could not detect these phosphorylated residues in the Tau aggregates, suggesting that the epitopes are either missing (residues not phosphorylated or cleaved of) or embedded inside the misfolding. Given the complexity of Tau post-translational modification, one cannot rule out that the aggregation may be mediated by some unidentified modification(s) of Tau. It is also possible that other factors, such as molecules that bind to Tau, may precipitate the aggregation.


Although the underlying mechanism is still unclear, the finding of Tau pathology in these mice establishes a causal link between the expression of amyloidogenic APP transgenes and a dysregulation of Tau assembly. The data also suggest a possibility that PTPσ depletion may suppress Tau aggregation by reducing amyloidogenic products of APP.


Malfunction of Tau is broadly recognized as a neurodegenerative marker since it indicates microtubule deterioration7. The constraining effect on Tau aggregation by genetic depletion of PTPσ thus provides additional evidence for the role of this receptor as a pivotal regulator of neuronal integrity.


PTPσ Deficiency Rescues Behavioral Deficits in AD Mouse Models.


Next, the question was assessed of whether the alleviation of neuropathologies by PTPσ depletion is accompanied with a rescue from AD relevant behavioral deficits. The most common symptoms of AD include short-term memory loss and apathy among the earliest, followed by spatial disorientation amid impairment of many cognitive functions as the dementia progresses. Using Y maze and novel object assays as surrogate models, these cognitive and psychiatric features were evaluated in the TgAPP-SwDI and TgAPP-SwInd mice.


The Y-maze assay, which allows mice to freely explore three identical arms, measures their short-term spatial memory. It is based on the natural tendency of mice to alternate arm exploration without repetitions. The performance is scored by the percentage of spontaneous alternations among total arm entries, and a higher score indicates better spatial navigation. Compared to the non-transgenic wild type mice within the colony, the APP-SwDI(+)PTPσ(+/+) mice show a clear deficit in their performance. Genetic depletion of PTPσ in the APP-SwDI(+)PTPσ(−/−) mice, however, unequivocally restores the cognitive performance back to the level of non-transgenic wild type mice (FIG. 13a, FIG. 14).


Apathy, the most common neuropsychiatric symptom reported among individuals with AD, is characterized by a loss of motivation and diminished attention to novelty, and has been increasingly adopted into early diagnosis of preclinical and early prodromal AD34-36. Many patients in early stage AD lose attention to novel aspects of their environment despite their ability to identify novel stimuli, suggesting an underlying defect in the circuitry responsible for further processing of the novel information34,35. As a key feature of apathy, such deficits in attention to novelty can be accessed by the “curiosity figures task” or the “oddball task” in patients34,35,37. These visual-based novelty encoding tasks are very similar to the novel object assay for rodents, which measures the interest of animals in a novel object (NO) when they are exposed simultaneously to a prefamiliarized object (FO). This assay was therefore used to test the attention to novelty in the APP transgenic mice. When mice are pre-trained to recognize the FO, their attention to novelty is then measured by the discrimination index denoted as the ratio of NO exploration to total object exploration (NO+FO), or alternatively, by the ratio of NO exploration to FO exploration. Whereas both ratios are commonly used, a combination of these assessments provides a more comprehensive evaluation of animal behavior. In this test, as indicated by both measurements, the expression of APP-SwDI transgene in the APP-SwDI(+)PTPσ(+/+) mice leads to a substantial decrease in NO exploration as compared to non-transgenic wild type mice (FIG. 11b, c; FIG. 15). Judging by their NO/FO ratios, it is evident that both the transgenic and non-transgenic groups are able to recognize and differentiate between the two objects (FIG. 15a, b). Thus, the reduced NO exploration by the APP-SwDI(+)PTPσ(+/+) mice may reflect a lack of interest in the NO or an inability to shift attention to the NO. Once again, this behavioral deficit is largely reversed by PTPσ deficiency in the APP-SwDI(+)PTPσ(−/−) mice (FIG. 13b, c; FIG. 15), consistent with previous observation of increased NO preference in the absence of PTPσ28.


To further verify the effects of PTPσ on these behavioral aspects, the TgAPP-SwInd mice were also tested using both assays, and similar results were observed. This confirms an improvement on both short-term spatial memory and attention to novelty upon genetic depletion of PTPσ (FIG. 16).


Discussion

The above data showed that β-amyloidosis and several downstream disease features are dependent on PTPσ in two mouse models of genetically inherited AD. This form of AD develops inevitably in people who carry gene mutations that promote amyloidogenic processing of APP and overproduction of Aβ. The data presented herein suggest that targeting PTPσ is a potential therapeutic approach that could overcome such dominant genetic driving forces to curtail AD progression. The advantage of this targeting strategy is that it suppresses Aβ accumulation without broadly affecting other major substrates of the β- and γ-secretases, thus predicting a more promising translational potential as compared to those in clinical trials that generically inhibit the secretases.


PTPσ was previously characterized as a neuronal receptor of the chondroitin sulfate- and heparan sulfate-proteoglycans (CSPGs and HSPGs)10,11. In response to these two classes of extracellular ligands, PTPσ functions as a “molecular switch” by regulating neuronal behavior in opposite manners8. The finding presented herein of a pivotal role for the proteoglycan sensor PTPσ in AD pathogenesis may therefore implicate an involvement of the perineuronal matrix in AD etiology.


More than 95% of AD cases are sporadic, which are not genetically inherited but likely result from insults to the brain that occurred earlier in life. AD risk factors, such as traumatic brain injury and cerebral ischemia38-41, have been shown to induce overproduction of Aβ in both human and rodents42-46, and speed up progression of this dementia in animal models47-49. However, what promotes the amyloidogenic processing of APP in these cases is still a missing piece of the puzzle in understanding the AD-causing effects of these notorious risk factors.


Coincidently, both traumatic brain injury and cerebral ischemia cause pronounced remodeling of the perineuronal microenvironment at lesion sites, marked by increased expression of CSPGs50-53, a major component of the perineuronal net that is upregulated during neuroinflammation and glial scar formation54-56. In the brains of AD patients, CSPGs were found associated with Aβ depositions, further suggesting an uncanny involvement of these proteoglycans in AD development57. On the other hand, analogues of heparan sulfate (HS, carbohydrate side chains of HSPGs that bind to PTPσ) were shown to inhibit BACE1 activity, suggesting their function in preventing Aβ overproduction58. After cerebral ischemia, however, the expression of Heparanase, an enzyme that degrades HS, was found markedly increased59. Collectively, these findings suggest a disrupted molecular balance between CSPGs and HSPGs in brains after lesion, which may ignite insidious signaling cascades preceding the onset of AD.


Further study could include investigation of a potential mechanism, whereby chronic CSPG upregulation or HSPG degradation in lesioned brains may sustain aberrant signaling through their neuronal sensor PTPσ, leading to biased processing of APP and a neurotoxic “Aβ cascade”. As such, altered signaling from PTPσ after traumatic brain injury and ischemic stroke may explain how these risk factors can trigger subsequent onset of AD. Restoring the integrity of brain microenvironment therefore could be essential in preventing AD for the population at risk.


Example 2: CS and HS Regulates APP Amyloidogenic Processing in Opposite Manners

CS and HS/heparin are two classes of PTPσ ligands in the perineuronal space that compete for binding to the same site on receptor PTPσ with similar affinities8. Increased CS/HS ratio is often found after brain injuries or ischemic stroke50-53,59, both of which are prominent risk factors for AD and alike neurodegenerative diseases.


These two classes of ligands were shown previously to oppositely regulate neuronal responses, such as neurite outgrowth, through their common receptor PTPσ. Whereas CS inhibits neurite outgrowth, HS/heparin promotes neurite outgrowth.


When tested in an in vitro assay for their effects on APP amyloidogenic processing, these PTPσ ligands again showed opposite effects. As in FIG. 17, incubation of cell membrane preparations extracted from fresh mouse brain homogenates with these PTPσ ligands results in an increased level of APP β-cleavage by CS, but a decreased level of APP β-cleavage by HS/heparin. Whereas CS levels are well documented to be upregulated after traumatic brain injury (TBI) in rats and mice, this study found increased APP-PTPσ binding accompanied with significantly enhanced level of APP β-cleavage product (CTFβ) in injured brains (FIG. 18). On the contrary, HS/heparin, which inhibits APP β-cleavage, effectively disrupts APP-PTPσ binding (FIG. 19). These data thus suggest that the molecular balance of PTPσ ligands CS and HS in the brain is important in regulating APP amyloidogenic processing, and that the promoting and suppressing effects on APP β-cleavage by CS and HS, respectively, are mediated by their control on APP-PTPσ binding.


Example 3: Defining Binding Regions on Human APP and PTPσ

Domain regions were subcloned from human APP695 (construct by Denis Selkoe and Tracy Yang labs purchased through Addgene.com) and PTPσ (constructs from Radu Aricescu lab). Recombinant APP and PTPσ proteins were tested in solid phase ELISA binding assays to define the binding regions on each partner. Neither E1 or E2 domain of APP interacts with PTPσ (data not shown), however the region in between these two APP domains (SEQ ID NO:1) appears to have high affinity with PTPσ IG1 domain (FIG. 20). The lysine residues (K67, 68, 70, 71) in PTPσ IG1 ligand binding site, which was shown to be responsible for CS and HS binding8,11,60 are also important for its interaction with APP, as mutation of these residues abolishes APP-PTPσ binding. Comparing APP binding strength of difference PTPσ fragments, it appears that inclusion of the fibronectin (FN) domains of PTPσ weakens the interaction with APP, likely due to folding of PTPσ that covers up the ligand binding site in its IG1 domain61. Full PTPσ extracellular domain nearly lost binding with APP SEQ ID NO:1, suggesting that factors triggering the unfold PTPσ are required for APP-PTPσ binding.


Sequences:


Sequences for the peptides used in Example 3 are provided in Tables 3, 4, and 5.









TABLE 3





Peptides derived from APP


















SEQ ID NO: 101
ADAEEDDSDVW







SEQ ID NO: 112
WGGADTDYADG







SEQ ID NO: 388
EDKVVEVAEEEEVA







SEQ ID NO: 139
VEEEEADDDED







SEQ ID NO: 151
EDGDEVEEEAE







SEQ ID NO: 157
EEEAEEPYEEA







SEQ ID NO: 251
EPYEEATERTTS







SEQ ID NO: 897
ESVEEVVRVPTTA







SEQ ID NO: 900
ATERTTSIATTTTTTTESVEEVVR

















TABLE 4





Peptides derived from PTPσ


















SEQ ID NO: 655
TWNKKGKKVNSQ







SEQ ID NO: 769
RIQPLRTPRDENV







SEQ ID NO: 898
KKGKK







SEQ ID NO: 899
RTPR

















TABLE 5





Membrane penetrating peptides


















SEQ ID NO: 895
GRKKRRQRRRPQ







SEQ ID NO: 896
RKKRRQRRRC










Unless defined otherwise, all technical and scientific terms used herein have the same meanings as commonly understood by one of skill in the art to which the disclosed invention belongs. Publications cited herein and the materials for which they are cited are specifically incorporated by reference.


Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.


REFERENCES



  • 1. Selkoe, D. J. Alzheimer's disease. Cold Spring Harbor perspectives in biology 3(2011).

  • 2. Yan, R. & Vassar, R. Targeting the beta secretase BACE1 for Alzheimer's disease therapy. The Lancet. Neurology 13, 319-329 (2014).

  • 3. Mikulca, J. A., et al. Potential novel targets for Alzheimer pharmacotherapy: II. Update on secretase inhibitors and related approaches. Journal of clinical pharmacy and therapeutics 39, 25-37 (2014).

  • 4. De Strooper, B. Lessons from a failed gamma-secretase Alzheimer trial. Cell 159, 721-726 (2014).

  • 5. Arriagada, P. V., Growdon, J. H., Hedley-Whyte, E. T. & Hyman, B. T. Neurofibrillary tangles but not senile plaques parallel duration and severity of Alzheimer's disease. Neurology 42, 631-639 (1992).

  • 6. Bouras, C., Hof P. R., Giannakopoulos, P., Michel, J. P. & Morrison, J. H. Regional distribution of neurofibrillary tangles and senile plaques in the cerebral cortex of elderly patients: a quantitative evaluation of a one-year autopsy population from a geriatric hospital. Cerebral cortex 4, 138-150 (1994).

  • 7. Wang, Y. & Mandelkow, E. Tau in physiology and pathology. Nature reviews. Neuroscience 17, 5-21 (2016).

  • 8. Coles, C. H., et al. Proteoglycan-specific molecular switch for RPTPsigma clustering and neuronal extension. Science 332, 484-488 (2011).

  • 9. Elchebly, M., et al. Neuroendocrine dysplasia in mice lacking protein tyrosine phosphatase sigma. Nature genetics 21, 330-333 (1999).

  • 10. Aricescu, A. R., McKinnell, I. W., Halfter, W. & Stoker, A. W. Heparan sulfate proteoglycans are ligands for receptor protein tyrosine phosphatase sigma. Molecular and cellular biology 22, 1881-1892 (2002).

  • 11. Shen, Y, et al. PTPsigma is a receptor for chondroitin sulfate proteoglycan, an inhibitor of neural regeneration. Science 326, 592-596 (2009).

  • 12. Wang, H., et al. Expression of receptor protein tyrosine phosphatase-sigma (RPTP-sigma) in the nervous system of the developing and adult rat. Journal of neuroscience research 41, 297-310 (1995).

  • 13. Yan, H., et al. A novel receptor tyrosine phosphatase-sigma that is highly expressed in the nervous system. The Journal of biological chemistry 268, 24880-24886 (1993).

  • 14. Chow, V. W., Mattson, M. P., Wong, P. C. & Gleichmann, M. An overview of APP processing enzymes and products. Neuromolecular medicine 12, 1-12 (2010).

  • 15. Nunan, J. & Small, D. H. Regulation of APP cleavage by alpha-, beta- and gamma-secretases. FEBS letters 483, 6-10 (2000).

  • 16. Estus, S., et al. Potentially amyloidogenic, carboxyl-terminal derivatives of the amyloid protein precursor. Science 255, 726-728 (1992).

  • 17. Davis, J., et al. Early-onset and robust cerebral microvascular accumulation of amyloid beta-protein in transgenic mice expressing low levels of a vasculotropic Dutch/Iowa mutant form of amyloid beta-protein precursor. The Journal of biological chemistry 279, 20296-20306 (2004).

  • 18. Mucke, L., et al. High-level neuronal expression of abeta 1-42 in wild-type human amyloid protein precursor transgenic mice: synaptotoxicity without plaque formation. The Journal of neuroscience: the official journal of the Society for Neuroscience 20, 4050-4058 (2000).

  • 19. Fleck, D., Garratt, A. N., Haass, C. & Willem, M. BACE1 dependent neuregulin processing: review. Current Alzheimer research 9, 178-183 (2012).

  • 20. Luo, X., et al. Cleavage of neuregulin-1 by BACE1 or ADAM10 protein produces differential effects on myelination. The Journal of biological chemistry 286, 23967-23974 (2011).

  • 21. Cheret, C., et al. Bace1 and Neuregulin-1 cooperate to control formation and maintenance of muscle spindles. The EMBO journal 32, 2015-2028 (2013).

  • 22. De Strooper, B., et al. A presenilin-1-dependent gamma-secretase-like protease mediates release of Notch intracellular domain. Nature 398, 518-522 (1999).

  • 23. Tian, Y, Bassit, B., Chau, D. & Li, Y. M. An APP inhibitory domain containing the Flemish mutation residue modulates gamma-secretase activity for Abeta production. Nature structural & molecular biology 17, 151-158 (2010).

  • 24. Zhang, Z., et al. Presenilins are required for gamma-secretase cleavage of beta-APP and transmembrane cleavage of Notch-1. Nature cell biology 2, 463-465 (2000).

  • 25. Glass, C. K., Saijo, K., Winner, B., Marchetto, M. C. & Gage, F. H. Mechanisms underlying inflammation in neurodegeneration. Cell 140, 918-934 (2010).

  • 26. DeWitt, D. A., Perry, G., Cohen, M., Doller, C. & Silver, J. Astrocytes regulate microglial phagocytosis of senile plaque cores of Alzheimer's disease. Experimental neurology 149, 329-340 (1998).

  • 27. Frederickson, R. C. Astroglia in Alzheimer's disease. Neurobiology of aging 13, 239-253 (1992).

  • 28. Horn, K. E., et al. Receptor protein tyrosine phosphatase sigma regulates synapse structure, function and plasticity. Journal of neurochemistry 122, 147-161 (2012).

  • 29. Tomidokoro, Y, et al. Abeta amyloidosis induces the initial stage of tau accumulation in APP(Sw) mice. Neuroscience letters 299, 169-172 (2001).

  • 30. Sturchler-Pierrat, C., et al. Two amyloid precursor protein transgenic mouse models with Alzheimer disease-like pathology. Proceedings of the National Academy of Sciences of the United States of America 94, 13287-13292 (1997).

  • 31. Rockenstein, E., Mallory, M., Mante, M., Sisk, A. & Masliaha, E. Early formation of mature amyloid-beta protein deposits in a mutant APP transgenic model depends on levels of Abeta(1-42). Journal of neuroscience research 66, 573-582 (2001).

  • 32. Holtnnan, D. M., et al. Tau: From research to clinical development. Alzheimer's & dementia: the journal of the Alzheimer's Association (2016).

  • 33. Morris, M., et al. Tau post-translational modifications in wild-type and human amyloid precursor protein transgenic mice. Nature neuroscience 18, 1183-1189 (2015).

  • 34. Daffier, K. R., et al. Pathophysiology underlying diminished attention to novel events in patients with early AD. Neurology 56, 1377-1383 (2001).

  • 35. Daffier, K. R., Mesulam, M. M., Cohen, L. G. & Scinto, L. F. Mechanisms underlying diminished novelty-seeking behavior in patients with probable Alzheimer's disease. Neuropsychiatry, neuropsychology, and behavioral neurology 12, 58-66 (1999).

  • 36. Marin, R. S., Biedrzycki, R. C. & Firinciogullari, S. Reliability and validity of the Apathy Evaluation Scale. Psychiatry research 38, 143-162 (1991).

  • 37. Kaufman, D. A., Bowers, D., Okun, M. S., Van Patten, R. & Perlstein, W. M. Apathy, Novelty Processing, and the P3 Potential in Parkinson's Disease. Frontiers in neurology 7, 95 (2016).

  • 38. Johnson, V. E., Stewart, W. & Smith, D. H. Traumatic brain injury and amyloid-beta pathology: a link to Alzheimer's disease? Nature reviews. Neuroscience 11, 361-370 (2010).

  • 39. Sivanandam, T. M. & Thakur, M. K. Traumatic brain injury: a risk factor for Alzheimer's disease. Neuroscience and biobehavioral reviews 36, 1376-1381 (2012).

  • 40. Kalaria, R. N. The role of cerebral ischemia in Alzheimer's disease. Neurobiology of aging 21, 321-330 (2000).

  • 41. Cole, S. L. & Vassar, R. Linking vascular disorders and Alzheimer's disease: potential involvement of BACE1. Neurobiology of aging 30, 1535-1544 (2009).

  • 42. Emmerling, M. R., et al. Traumatic brain injury elevates the Alzheimer's amyloid peptide A beta 42 in human CSF. A possible role for nerve cell injury. Annals of the New York Academy of Sciences 903, 118-122 (2000).

  • 43. Olsson, A., et al. Marked increase of beta-amyloid(1-42) and amyloid precursor protein in ventricular cerebrospinal fluid after severe traumatic brain injury. Journal of neurology 251, 870-876 (2004).

  • 44. Loane, D. J., et al. Amyloid precursor protein secretases as therapeutic targets for traumatic brain injury. Nature medicine 15, 377-379 (2009).

  • 45. Pluta, R., Furmaga-Jablonska, W., Maciejewski, R., Ulamek-Koziol, M. & Jablonski, M. Brain ischemia activates beta- and gamma-secretase cleavage of amyloid precursor protein: significance in sporadic Alzheimer's disease. Molecular neurobiology 47, 425-434 (2013).

  • 46. Washington, P. M., et al. The effect of injury severity on behavior: a phenotypic study of cognitive and emotional deficits after mild, moderate, and severe controlled cortical impact injury in mice. Journal of neurotrauma 29, 2283-2296 (2012).

  • 47. Kokiko-Cochran, O., et al. Altered Neuroinflammation and Behavior after Traumatic Brain Injury in a Mouse Model of Alzheimer's Disease. Journal of neurotrauma (2015).

  • 48. Tajiri, N., Kellogg S. L., Shimizu, T., Arendash, G. W. & Borlongan, C. V. Traumatic brain injury precipitates cognitive impairment and extracellular Abeta aggregation in Alzheimer's disease transgenic mice. PloS one 8, e78851 (2013).

  • 49. Watanabe, T., Takasaki, K., Yamagata, N., Fujiwara, M. & Iwasaki, K. Facilitation of memory impairment and cholinergic disturbance in a mouse model of Alzheimer's disease by mild ischemic burden. Neuroscience letters 536, 74-79 (2013).

  • 50. Properzi, F., et al. Chondroitin 6-sulphate synthesis is up-regulated in injured CNS, induced by injury-related cytokines and enhanced in axon-growth inhibitory glia. The European journal of neuroscience 21, 378-390 (2005).

  • 51. Yi, J. H., et al. Alterations in sulfated chondroitin glycosaminoglycans following controlled cortical impact injury in mice. The Journal of comparative neurology 520, 3295-3313 (2012).

  • 52. Hill, J. J., Jin, K., Mao, X. O., Xie, L. & Greenberg, D. A. Intracerebral chondroitinase ABC and heparan sulfate proteoglycan glypican improve outcome from chronic stroke in rats. Proceedings of the National Academy of Sciences of the United States of America 109, 9155-9160 (2012).

  • 53. Huang, L., et al. Glial scar formation occurs in the human brain after ischemic stroke. International journal of medical sciences 11, 344-348 (2014).

  • 54. Celio, M. R. & Blumcke, I. Perineuronal nets—a specialized form of extracellular matrix in the adult nervous system. Brain research. Brain research reviews 19, 128-145 (1994).

  • 55. Cregg, J. M., et al. Functional regeneration beyond the glial scar. Experimental neurology 253, 197-207 (2014).

  • 56. Soleman, S., Filippov, M. A., Dityatev, A. & Fawcett, J. W. Targeting the neural extracellular matrix in neurological disorders. Neuroscience 253, 194-213 (2013).

  • 57. DeWitt, D. A., Silver, J., Canning, D. R. & Perry, G. Chondroitin sulfate proteoglycans are associated with the lesions of Alzheimer's disease. Experimental neurology 121, 149-152 (1993).

  • 58. Patey, S. J., Edwards, E. A., Yates, E. A. & Turnbull, J. E. Heparin derivatives as inhibitors of BACE-1, the Alzheimer's beta-secretase, with reduced activity against factor Xa and other proteases. Journal of medicinal chemistry 49, 6129-6132 (2006).

  • 59. Li, J., et al. Expression of heparanase in vascular cells and astrocytes of the mouse brain after focal cerebral ischemia. Brain research 1433, 137-144 (2012).

  • 60. Sajnani-Perez, G., Chilton, J. K., Aricescu, A. R., Haj, F. & Stoker, A. W. Isoform-specific binding of the tyrosine phosphatase PTPsigma to a ligand in developing muscle. Molecular and cellular neurosciences 22, 37-48 (2003).

  • 61. Coles, C. H., et al. Structural basis for extracellular cis and trans RPTPsigma signal competition in synaptogenesis. Nature communications 5, 5209 (2014).


Claims
  • 1. A peptide for treating or preventing a neurodegenerative disorder, the peptide comprising; a decoy fragment of Amyloid Precursor Protein (APP), a decoy fragment of Receptor Protein Tyrosine Phosphatase Sigma (PTPσ), or a combination thereof, anda blood brain barrier penetrating sequence.
  • 2. The peptide of claim 1, wherein the decoy fragment of APP is a peptide comprising at least 5 consecutive amino acids of SEQ ID NO:1.
  • 3. The peptide of claim 2, wherein the decoy fragment of APP is a peptide comprising at least 10 consecutive amino acids of SEQ ID NO:1.
  • 4. The peptide of claim 1, wherein the decoy fragment of APP comprises an amino acid sequence selected from the group consisting of SEQ ID NO:88, SEQ ID NO:91, SEQ ID NO:101, SEQ ID NO:112, SEQ ID NO:139, SEQ ID NO:151, SEQ ID NO:157, SEQ ID NO:251, SEQ ID NO:897, SEQ ID NO: 900.
  • 5. The peptide of claim 1, wherein the decoy fragment of PTPσ is a peptide comprising at least 4 consecutive amino acids of SEQ ID NO:442.
  • 6. The peptide of claim 5, wherein the decoy fragment of PTPσ is a peptide comprising at least 10 consecutive amino acids of SEQ ID NO:442.
  • 7. The peptide of claim 5, wherein the decoy fragment of PTPσ comprises the amino acid sequence SEQ ID NO:898, SEQ ID NO:899. SEQ ID NO:655, or SEQ ID NO:769.
  • 8. The peptide of claim 1, wherein the blood brain barrier penetrating sequence comprises amino acid sequence SEQ ID NO: 880, SEQ ID NO: 883, SEQ ID NO: 888, SEQ ID NO: 894, SEQ ID NO: 895, SEQ ID NO: 896.
  • 9. The peptide of claim 1, wherein the peptide is cyclic.
  • 10. A composition, comprising the peptide of claim 1 and further comprising a pharmaceutically acceptable excipient.
  • 11. An antibody or an antibody fragment against APP or PTPσ for treating or preventing a neurodegenerative disorder, wherein the antibody or antibody fragment binds an epitope on APP or an epitope on PTPσ.
  • 12. The antibody or antibody fragment of claim 11, wherein the epitope on APP is a peptide sequence between the E1 and E2 domains of APP.
  • 13. The antibody or antibody fragment of claim 11, wherein the epitope on PTPσ is a peptide sequence on the PTPσ IG1 domain.
  • 14. The antibody or antibody fragment of claim 11, wherein the epitope on PTPσ is the entire PTPσ IG1 domain or SEQ ID NO:442.
  • 15. The antibody or antibody fragment of claim 11, further comprising a pharmaceutically acceptable excipient.
  • 16. One or more compounds or enzymes for treating or preventing a neurodegenerative disorder, wherein the compound or enzyme restores the physiological molecular balance of chondroitin sulfate (CS) and heparan sulfate (HS) in the brain.
  • 17. The one or more compounds or enzymes of claim 16, wherein the compound or enzyme is an analog of heparin, an analog of HS, a mimetic of heparin, a mimetic of HS, an inhibitor of heparanase, chondroitinase ABC (ChABC), or a combination thereof.
  • 18. The one or more compounds or enzymes of claim 16, wherein the compound is an inhibitor of heparanase.
  • 19. The one or more compounds or enzymes of claim 16, wherein the compound is an analog or mimetic of heparin or HS.
  • 20. The compound or enzyme of claim 16, wherein the compound or enzyme is ChABC.
  • 21. The compound or enzyme of claim 17, further comprising a pharmaceutically acceptable excipient.
  • 22.-34. (canceled)
CROSS REFERENCE TO RELATED APPLICATIONS

This application claims the benefit of U.S. Provisional Application No. 62/335,159, filed May 12, 2016, which is hereby incorporated by reference in its entirety for all purposes.

PCT Information
Filing Document Filing Date Country Kind
PCT/US2017/032387 5/12/2017 WO 00
Provisional Applications (1)
Number Date Country
62335159 May 2016 US