119 results on '"Brunden KR"'
Search Results
2. Brain-penetrant, orally bioavailable microtubule-stabilizing small molecules as potential candidates for the treatment of Alzheimer's disease and related tauopathies
- Author
-
Ballatore, C, Lou, K, Hoye, A, Gay, B, Cornec, A-S, James, MJ, Yao, Y, Hyde, E, Trojanowski, JQ, Lee, VM-Y, Smith, AB, and Brunden, KR
- Subjects
Mice ,Pyrimidines ,Tauopathies ,Molecular Structure ,Alzheimer Disease ,Brain ,Animals ,Humans ,Administration, Oral ,Biological Availability ,Microtubules ,Cell Line - Abstract
Microtubule (MT) stabilizing drugs hold promise as potential treatments for Alzheimer's disease (AD) and related tauopathies. However, thus far epothilone D has been the only brain-penetrant MT-stabilizer to be evaluated in tau transgenic mice and in AD patients. Furthermore, this natural product exhibits potential deficiencies as a drug candidate, including an intravenous route of administration and the inhibition of the P-glycoprotein (Pgp) transporter. Thus, the identification of alternative CNS-active MT-stabilizing agents that lack these potential limitations is of interest. Toward this objective, we have evaluated representative compounds from known classes of non-naturally occurring MT-stabilizing small molecules. This led to the identification of selected triazolopyrimidines and phenylpyrimidines that are orally bioavailable and brain-penetrant without disruption of Pgp function. Pharmacodynamic studies confirmed that representative compounds from these series enhance MT-stabilization in the brains of wild-type mice. Thus, these classes of MT-stabilizers hold promise for the development of orally active, CNS-directed MT-stabilizing therapies.
- Published
- 2014
- Full Text
- View/download PDF
3. Evidence that non-fibrillar tau causes pathology linked to neurodegeneration and behavioral impairments.
- Author
-
Brunden KR, Trojanowski JQ, Lee VM, Brunden, Kurt R, Trojanowski, John Q, and Lee, Virginia M-Y
- Abstract
The discovery that mutations within the tau gene lead to frontotemporal dementia with Parkinsonism linked to chromosome 17 (FTDP-17) provided direct evidence that tau alterations can lead to neurodegenerative disease. While the presence of tau fibrils and tangles is a common feature of all tauopathies, including Alzheimer's disease (AD), data are emerging from biochemical, cell-based and transgenic mouse studies which suggest that a pre-fibrillar form of pathological tau may play a key role in eliciting central nervous system neurodegeneration and behavioral impairments. Herein we review recent findings that implicate diffusible tau pathology in the onset of neurodegeneration, and discuss the implications of these findings as they relate to tau tangles and possible therapeutic strategies for the treatment of AD and related tauopathies. [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
4. Targeting Endogenous Tau in Seeded Tauopathy Models Inhibits Tau Spread.
- Author
-
Jang E, Hoxha K, Mozier D, Insana A, Farber E, Changolkar L, Zhang B, Chio TI, Crowe A, Chen R, Mercken M, Lee EB, Luk KC, Brunden KR, Lee VM, and Xu H
- Subjects
- Animals, Mice, Humans, Female, Neurons metabolism, Disease Models, Animal, Mice, Inbred C57BL, Cells, Cultured, Mice, Transgenic, Blood-Brain Barrier metabolism, Alzheimer Disease metabolism, Alzheimer Disease pathology, Antibodies pharmacology, tau Proteins metabolism, Tauopathies metabolism, Tauopathies pathology, Tauopathies drug therapy
- Abstract
The transmission of tau pathology has been proposed as one of the major mechanisms for the spatiotemporal spreading of tau pathology in neurodegenerative diseases. Over the last decade, studies have demonstrated that targeting total or pathological tau using tau antibodies can mitigate the development of tau pathology in tauopathy or Alzheimer's disease (AD) mouse models, and multiple tau immunotherapy agents have progressed to clinical trials. Tau antibodies are believed to inhibit the internalization of pathologic seeds and/or block seed elongation after seed internalization. To further address the mechanism of tau antibody inhibition of pathological spread, we conducted immunotherapy studies in mouse primary neurons and wild-type mice (females) seeded with AD patient-derived tau to induce the formation and spreading of tau pathology. Notably, we evaluated the effect of a mouse tau-specific antibody (mTau8) which does not interact with AD-tau seeds in these models. Our results show that mTau8 crosses the blood-brain barrier at levels similar to other antibodies and effectively decreases AD-tau-seeded tau pathology in vitro and in vivo. Importantly, our data suggest that mTau8 binds to endogenous intraneuronal mouse tau, thereby inhibiting the elongation of internalized tau seeds. These findings provide valuable insights into the possible mechanism underlying antibody-based therapies for treating tauopathies., Competing Interests: The authors declare no competing financial interests., (Copyright © 2024 the authors.)
- Published
- 2024
- Full Text
- View/download PDF
5. A small-molecule microtubule-stabilizing agent safely reduces Aβ plaque and tau pathology in transgenic mouse models of Alzheimer's disease.
- Author
-
Yao Y, Muench M, Alle T, Zhang B, Lucero B, Perez-Tremble R, McGrosso D, Newman M, Gonzalez DJ, Lee VM, Ballatore C, and Brunden KR
- Subjects
- Animals, Mice, Brain drug effects, Brain pathology, Brain metabolism, Tauopathies drug therapy, Tauopathies pathology, Humans, Tubulin Modulators pharmacology, Tubulin Modulators therapeutic use, Amyloid beta-Peptides metabolism, Alzheimer Disease drug therapy, Alzheimer Disease pathology, Mice, Transgenic, Disease Models, Animal, Plaque, Amyloid drug therapy, Plaque, Amyloid pathology, tau Proteins metabolism, Microtubules drug effects, Microtubules metabolism
- Abstract
Introduction: Intraneuronal inclusions composed of tau protein are found in Alzheimer's disease (AD) and other tauopathies. Tau normally binds microtubules (MTs), and its disengagement from MTs and misfolding in AD is thought to result in MT abnormalities. We previously identified triazolopyrimidine-containing MT-stabilizing compounds that provided benefit in AD mouse models and herein describe the characterization and efficacy testing of an optimized candidate, CNDR-51997., Methods: CNDR-51997 underwent pharmacokinetic, pharmacodynamic, safety pharmacology, and mouse tolerability testing. In addition, the compound was examined for efficacy in 5XFAD amyloid beta (Aβ) plaque mice and PS19 tauopathy mice., Results: CNDR-51997 significantly reduced Aβ plaques in 5XFAD mice and tau pathology in PS19 mice, with the latter also showing attenuated axonal dystrophy and gliosis. CNDR-51997 was well tolerated at doses that exceeded efficacy doses, with a good safety pharmacology profile., Discussion: CNDR-51997 may be a candidate for advancement as a potential therapeutic agent for AD and/or other tauopathies. Highlights There is evidence of microtubule alterations (MT) in Alzheimer's disease (AD) brain and in mouse models of AD pathology. Intermittent dosing with an optimized, brain-penetrant MT-stabilizing small-molecule, CNDR-51997, reduced both Aβ plaque and tau inclusion pathology in established mouse models of AD. CNDR-51997 attenuated axonal dystrophy and gliosis in a tauopathy mouse model, with a strong trend toward reduced hippocampal neuron loss. CNDR-51997 is well tolerated in mice at doses that are meaningfully greater than required for efficacy in AD mouse models, and the compound has a good safety pharmacology profile., (© 2024 The Authors. Alzheimer's & Dementia published by Wiley Periodicals LLC on behalf of Alzheimer's Association.)
- Published
- 2024
- Full Text
- View/download PDF
6. Design, Synthesis, and Evaluation of An Anti-trypanosomal [1,2,4]Triazolo[1,5-a]pyrimidine Probe for Photoaffinity Labeling Studies.
- Author
-
Lucero B, Francisco KR, Varricchio C, Liu LJ, Yao Y, Brancale A, Brunden KR, Caffrey CR, and Ballatore C
- Subjects
- Animals, Humans, Tubulin metabolism, HEK293 Cells, Proteomics, Pyrimidines chemistry, Mammals metabolism, Trypanosomiasis, African drug therapy, Trypanosoma brucei brucei metabolism, Trypanocidal Agents chemistry
- Abstract
Studies have shown that depending on the substitution pattern, microtubule (MT)-targeting 1,2,4-triazolo[1,5-a]pyrimidines (TPDs) can produce different cellular responses in mammalian cells that may be due to these compounds interacting with distinct binding sites within the MT structure. Selected TPDs are also potently bioactive against the causative agent of human African trypanosomiasis, Trypanosoma brucei, both in vitro and in vivo. So far, however, there has been no direct evidence of tubulin engagement by these TPDs in T. brucei. Therefore, to enable further investigation of anti-trypanosomal TPDs, a TPD derivative amenable to photoaffinity labeling (PAL) was designed, synthesized, and evaluated in PAL experiments using HEK293 cells and T. brucei. The data arising confirmed specific labeling of T. brucei tubulin. In addition, proteomic data revealed differences in the labeling profiles of tubulin between HEK293 and T. brucei, suggesting structural differences between the TPD binding site(s) in mammalian and trypanosomal tubulin., (© 2024 The Authors. ChemMedChem published by Wiley-VCH GmbH.)
- Published
- 2024
- Full Text
- View/download PDF
7. MSUT2 regulates tau spreading via adenosinergic signaling mediated ASAP1 pathway in neurons.
- Author
-
Xu H, Qiu Q, Hu P, Hoxha K, Jang E, O'Reilly M, Kim C, He Z, Marotta N, Changolkar L, Zhang B, Wu H, Schellenberg GD, Kraemer B, Luk KC, Lee EB, Trojanowski JQ, Brunden KR, and Lee VM
- Subjects
- Mice, Humans, Animals, Brain pathology, tau Proteins metabolism, Neurons pathology, Mice, Transgenic, Mammals metabolism, Adaptor Proteins, Signal Transducing metabolism, Tauopathies pathology, Alzheimer Disease pathology
- Abstract
Inclusions comprised of microtubule-associated protein tau (tau) are implicated in a group of neurodegenerative diseases, collectively known as tauopathies, that include Alzheimer's disease (AD). The spreading of misfolded tau "seeds" along neuronal networks is thought to play a crucial role in the progression of tau pathology. Consequently, restricting the release or uptake of tau seeds may inhibit the spread of tau pathology and potentially halt the advancement of the disease. Previous studies have demonstrated that the Mammalian Suppressor of Tauopathy 2 (MSUT2), an RNA binding protein, modulates tau pathogenesis in a transgenic mouse model. In this study, we investigated the impact of MSUT2 on tau pathogenesis using tau seeding models. Our findings indicate that the loss of MSUT2 mitigates human tau seed-induced pathology in neuron cultures and mouse models. In addition, MSUT2 regulates many gene transcripts, including the Adenosine Receptor 1 (A1AR), and we show that down regulation or inhibition of A1AR modulates the activity of the "ArfGAP with SH3 Domain, Ankyrin Repeat, and PH Domain 1 protein" (ASAP1), thereby influencing the internalization of pathogenic tau seeds into neurons resulting in reduction of tau pathology., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
8. Structure-Activity Relationships, Tolerability and Efficacy of Microtubule-Active 1,2,4-Triazolo[1,5-a]pyrimidines as Potential Candidates to Treat Human African Trypanosomiasis.
- Author
-
Monti L, Liu LJ, Varricchio C, Lucero B, Alle T, Yang W, Bem-Shalom I, Gilson M, Brunden KR, Brancale A, Caffrey CR, and Ballatore C
- Subjects
- Humans, Mice, Animals, Tubulin metabolism, Pyrimidines pharmacology, Pyrimidines therapeutic use, Pyrimidines chemistry, Microtubules metabolism, Structure-Activity Relationship, Mammals metabolism, Trypanosomiasis, African drug therapy, Trypanosoma brucei brucei metabolism, Trypanocidal Agents pharmacology, Trypanocidal Agents therapeutic use, Trypanocidal Agents chemistry
- Abstract
Tubulin and microtubules (MTs) are potential protein targets to treat parasitic infections and our previous studies have shown that the triazolopyrimidine (TPD) class of MT-active compounds hold promise as antitrypanosomal agents. MT-targeting TPDs include structurally related but functionally diverse congeners that interact with mammalian tubulin at either one or two distinct interfacial binding sites; namely, the seventh and vinca sites, which are found within or between α,β-tubulin heterodimers, respectively. Evaluation of the activity of 123 TPD congeners against cultured Trypanosoma brucei enabled a robust quantitative structure-activity relationship (QSAR) model and the prioritization of two congeners for in vivo pharmacokinetics (PK), tolerability and efficacy studies. Treatment of T. brucei-infected mice with tolerable doses of TPDs significantly decreased blood parasitemia within 24 h. Further, two once-weekly doses at 10 mg/kg of a candidate TPD significantly extended the survival of infected mice relative to infected animals treated with vehicle. Further optimization of dosing and/or the dosing schedule of these CNS-active TPDs may provide alternative treatments for human African trypanosomiasis., (© 2023 The Authors. ChemMedChem published by Wiley-VCH GmbH.)
- Published
- 2023
- Full Text
- View/download PDF
9. Preclinical characterization and IND-enabling safety studies for PNT001, an antibody that recognizes cis-pT231 tau.
- Author
-
Foster K, Manca M, McClure K, Koivula P, Trojanowski JQ, Havas D, Chancellor S, Goldstein L, Brunden KR, Kraus A, and Ahlijanian MK
- Subjects
- Humans, Mice, Animals, Brain metabolism, Mice, Transgenic, Neurofibrillary Tangles, Antibodies, Monoclonal, Humanized, tau Proteins metabolism, Tauopathies
- Abstract
Background: The cis-conformer of tau phosphorylated at threonine-231 (cis-pT231 tau) is hypothesized to contribute to tauopathies. PNT001 is a humanized, monoclonal antibody that recognizes cis-pT231 tau. PNT001 was characterized to assess clinical development readiness., Methods: Affinity and selectivity were assessed by surface plasmon resonance and enzyme-linked immunosorbent assay. Immunohistochemistry (IHC) was performed with brain sections from human tauopathy patients and controls. Real-time quaking-induced conversion (RT-QuIC) was used to assess whether PNT001 reduced tau seeds from Tg4510 transgenic mouse brain. Murine PNT001 was evaluated in vivo in the Tg4510 mouse., Results: The affinity of PNT001 for a cis-pT231 peptide was 0.3 to 3 nM. IHC revealed neurofibrillary tangle-like structures in tauopathy patients with no detectable staining in controls. Incubation of Tg4510 brain homogenates with PNT001 lowered seeding in RT-QuIC. Multiple endpoints were improved in the Tg4510 mouse. No adverse findings attributable to PNT001 were detected in Good Laboratory Practice safety studies., Discussion: The data support clinical development of PNT001 in human tauopathies., (© 2023 Pinteon Therapeutics, Inc and The Authors. Alzheimer's & Dementia published by Wiley Periodicals LLC on behalf of Alzheimer's Association.)
- Published
- 2023
- Full Text
- View/download PDF
10. A microtubule stabilizer ameliorates protein pathogenesis and neurodegeneration in mouse models of repetitive traumatic brain injury.
- Author
-
Zhao X, Zeng W, Xu H, Sun Z, Hu Y, Peng B, McBride JD, Duan J, Deng J, Zhang B, Kim SJ, Zoll B, Saito T, Sasaguri H, Saido TC, Ballatore C, Yao H, Wang Z, Trojanowski JQ, Brunden KR, Lee VM, and He Z
- Subjects
- Animals, Mice, Microtubules, DNA-Binding Proteins, Brain, Disease Models, Animal, Excipients, Mice, Transgenic, Brain Injuries, Traumatic, Alzheimer Disease
- Abstract
Tau pathogenesis is a hallmark of many neurodegenerative diseases, including Alzheimer's disease (AD). Although the events leading to initial tau misfolding and subsequent tau spreading in patient brains are largely unknown, traumatic brain injury (TBI) may be a risk factor for tau-mediated neurodegeneration. Using a repetitive TBI (rTBI) paradigm, we report that rTBI induced somatic accumulation of phosphorylated and misfolded tau, as well as neurodegeneration across multiple brain areas in 7-month-old tau transgenic PS19 mice but not wild-type (WT) mice. rTBI accelerated somatic tau pathology in younger PS19 mice and WT mice only after inoculation with tau preformed fibrils and AD brain-derived pathological tau (AD-tau), respectively, suggesting that tau seeds are needed for rTBI-induced somatic tau pathology. rTBI further disrupted axonal microtubules and induced punctate tau and TAR DNA binding protein 43 (TDP-43) pathology in the optic tracts of WT mice. These changes in the optic tract were associated with a decline of visual function. Treatment with a brain-penetrant microtubule-stabilizing molecule reduced rTBI-induced tau, TDP-43 pathogenesis, and neurodegeneration in the optic tract as well as visual dysfunction. Treatment with the microtubule stabilizer also alleviated rTBI-induced tau pathology in the cortices of AD-tau-inoculated WT mice. These results indicate that rTBI facilitates abnormal microtubule organization, pathological tau formation, and neurodegeneration and suggest microtubule stabilization as a potential therapeutic avenue for TBI-induced neurodegeneration.
- Published
- 2023
- Full Text
- View/download PDF
11. Identification of small molecules and related targets that modulate tau pathology in a seeded primary neuron model.
- Author
-
Gibbons GS, Gould H, Lee VM, Crowe A, and Brunden KR
- Subjects
- Animals, Humans, Rats, Amyloid beta-Peptides metabolism, Amyloid beta-Protein Precursor metabolism, Aspartic Acid Endopeptidases metabolism, Alzheimer Disease metabolism, Amyloid Precursor Protein Secretases metabolism, Neurons metabolism, tau Proteins genetics, tau Proteins metabolism
- Abstract
Alzheimer's disease (AD) is characterized by the presence of tau protein inclusions and amyloid beta (Aβ) plaques in the brain, with Aβ peptides generated by cleavage of the amyloid precursor protein (APP) by BACE1 and γ-secretase. We previously described a primary rat neuron assay in which tau inclusions form from endogenous rat tau after seeding cells with insoluble tau isolated from the human AD brain. Here, we used this assay to screen an annotated library of ∼8700 biologically active small molecules for their ability to reduce immuno-stained neuronal tau inclusions. Compounds causing ≥30% inhibition of tau aggregates with <25% loss of DAPI-positive cell nuclei underwent further confirmation testing and assessment of neurotoxicity, and non-neurotoxic hits were subsequently analyzed for inhibitory activity in an orthogonal ELISA that quantified multimeric rat tau species. Of the 173 compounds meeting all criteria, a subset of 55 inhibitors underwent concentration-response testing and 46 elicited a concentration-dependent reduction of neuronal tau inclusions that were distinct from measures of toxicity. Among the confirmed inhibitors of tau pathology were BACE1 inhibitors, several of which, along with γ-secretase inhibitors/modulators, caused a concentration-dependent lowering of neuronal tau inclusions and a reduction of insoluble tau by immunoblotting, although they did not decrease soluble phosphorylated tau species. In conclusion, we have identified a diverse set of small molecules and related targets that reduce neuronal tau inclusions. Notably, these include BACE1 and γ-secretase inhibitors, suggesting that a cleavage product from a shared substrate, such as APP, might affect tau pathology., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
12. Structure-Activity Relationships, Tolerability and Efficacy of Microtubule-Active 1,2,4-Triazolo[1,5- a ]pyrimidines as Potential Candidates to Treat Human African Trypanosomiasis.
- Author
-
Monti L, Liu LJ, Varricchio C, Lucero B, Alle T, Yang W, Bem-Shalom I, Gilson M, Brunden KR, Brancale A, Caffrey CR, and Ballatore C
- Abstract
Tubulin and microtubules (MTs) are potential protein targets to treat parasitic infections and our previous studies have shown that the triazolopyrimidine (TPD) class of MT- active compounds hold promise as antitrypanosomal agents. MT-targeting TPDs include structurally related but functionally diverse congeners that interact with mammalian tubulin at either one or two distinct interfacial binding sites; namely, the seventh and vinca sites, which are found within or between α,β-tubulin heterodimers, respectively. Evaluation of the activity of 123 TPD congeners against cultured Trypanosoma brucei enabled a robust quantitative structure-activity relationship (QSAR) model and the prioritization of two congeners for in vivo pharmacokinetics (PK), tolerability and efficacy studies. Treatment of T. brucei -infected mice with tolerable doses of TPDs 3 and 4 significantly decreased blood parasitemia within 24 h. Further, two once-weekly doses of 4 at 10 mg/kg significantly extended the survival of infected mice relative to infected animals treated with vehicle. Further optimization of dosing and/or the dosing schedule of these CNS-active TPDs may provide alternative treatments for human African trypanosomiasis.
- Published
- 2023
- Full Text
- View/download PDF
13. Abundant copathologies of polyglucosan bodies, frontotemporal lobar degeneration with TDP-43 inclusions and ageing-related tau astrogliopathy in a family with a GBE1 mutation.
- Author
-
Uemura MT, Suh ER, Robinson JL, Brunden KR, Grossman M, Irwin DJ, Lee VM, Trojanowski JQ, Lee EB, and Van Deerlin VM
- Subjects
- Brain pathology, Glycogen Storage Disease, Amyloid beta-Peptides metabolism, DNA-Binding Proteins metabolism, Mutation, Humans, Nervous System Diseases, tau Proteins metabolism, Frontotemporal Dementia pathology, 1,4-alpha-Glucan Branching Enzyme genetics, 1,4-alpha-Glucan Branching Enzyme metabolism, Glycogen Debranching Enzyme System genetics, Glycogen Debranching Enzyme System metabolism, Frontotemporal Lobar Degeneration pathology
- Abstract
Aims: Adult polyglucosan body disease (APBD) is a progressive neurogenetic disorder caused by 1,4-alpha-glucan branching enzyme 1 (GBE1) mutation with an accumulation of polyglucosan bodies (PBs) in the central and peripheral nervous systems as a pathological hallmark. Here, we report two siblings in a family with a GBE1 mutation with prominent frontotemporal lobar degeneration with TAR DNA-binding protein 43 (FTLD-TDP) and ageing-related tau astrogliopathy (ARTAG) copathologies with PBs in the central nervous system., Methods: Whole-genome sequencing (WGS) followed by Sanger sequencing (SS) was performed on three affected and two unaffected siblings in a pedigree diagnosed with familial frontotemporal dementia. Out of the affected siblings, autopsies were conducted on two cases, and brain samples were used for biochemical and histological analyses. Brain sections were stained with haematoxylin and eosin and immunostained with antibodies against ubiquitin, tau, amyloid β, α-synuclein, TDP-43 and fused in sarcoma (FUS)., Results: A novel single nucleotide deletion in GBE1, c.1280delG, was identified, which is predicted to result in a reading frameshift, p.Gly427Glufs*9. This variant segregated with disease in the family, is absent from population databases and is predicted to cause loss of function, a known genetic mechanism for APBD. The affected siblings showed a greater than 50% decrease in GBE protein levels. Immunohistochemical analysis revealed widespread FTLD-TDP (type A) and ARTAG pathologies as well as PBs in the brains of two affected siblings for whom an autopsy was performed., Conclusions: This is the first report of a family with several individuals with a FTD clinical phenotype and underlying copathologies of APBD, FTLD-TDP and ARTAG with a segregating GBE1 loss-of-function mutation in affected siblings. The finding of copathologies of APBD and FTLD-TDP suggests these processes may share a disease mechanism resulting from this GBE1 mutation., (© 2022 British Neuropathological Society.)
- Published
- 2023
- Full Text
- View/download PDF
14. Microtubule-Stabilizing 1,2,4-Triazolo[1,5- a ]pyrimidines as Candidate Therapeutics for Neurodegenerative Disease: Matched Molecular Pair Analyses and Computational Studies Reveal New Structure-Activity Insights.
- Author
-
Alle T, Varricchio C, Yao Y, Lucero B, Nzou G, Demuro S, Muench M, Vuong KD, Oukoloff K, Cornec AS, Francisco KR, Caffrey CR, Lee VM, Smith AB 3rd, Brancale A, Brunden KR, and Ballatore C
- Subjects
- Humans, Pyrimidines chemistry, Microtubules metabolism, Tubulin metabolism, Structure-Activity Relationship, Neurodegenerative Diseases drug therapy, Neurodegenerative Diseases metabolism, Alzheimer Disease drug therapy, Alzheimer Disease metabolism
- Abstract
Microtubule (MT)-stabilizing 1,2,4-triazolo[1,5- a ]pyrimidines (TPDs) hold promise as candidate therapeutics for Alzheimer's disease (AD) and other neurodegenerative conditions. However, depending on the choice of substituents around the TPD core, these compounds can elicit markedly different cellular phenotypes that likely arise from the interaction of TPD congeners with either one or two spatially distinct binding sites within tubulin heterodimers ( i.e. , the seventh site and the vinca site). In the present study, we report the design, synthesis, and evaluation of a series of new TPD congeners, as well as matched molecular pair analyses and computational studies, that further elucidate the structure-activity relationships of MT-active TPDs. These studies led to the identification of novel MT-normalizing TPD candidates that exhibit favorable ADME-PK, including brain penetration and oral bioavailability, as well as brain pharmacodynamic activity.
- Published
- 2023
- Full Text
- View/download PDF
15. Distinct characteristics of limbic-predominant age-related TDP-43 encephalopathy in Lewy body disease.
- Author
-
Uemura MT, Robinson JL, Cousins KAQ, Tropea TF, Kargilis DC, McBride JD, Suh E, Xie SX, Xu Y, Porta S, Uemura N, Van Deerlin VM, Wolk DA, Irwin DJ, Brunden KR, Lee VM, Lee EB, and Trojanowski JQ
- Subjects
- Aged, Aged, 80 and over, Alzheimer Disease complications, Alzheimer Disease genetics, Alzheimer Disease pathology, Female, Humans, Lewy Body Disease complications, Lewy Body Disease genetics, Male, Middle Aged, TDP-43 Proteinopathies complications, TDP-43 Proteinopathies genetics, Aging pathology, Brain pathology, Lewy Body Disease pathology, TDP-43 Proteinopathies pathology
- Abstract
Limbic-predominant age-related TDP-43 encephalopathy (LATE) is characterized by the accumulation of TAR-DNA-binding protein 43 (TDP-43) aggregates in older adults. LATE coexists with Lewy body disease (LBD) as well as other neuropathological changes including Alzheimer's disease (AD). We aimed to identify the pathological, clinical, and genetic characteristics of LATE in LBD (LATE-LBD) by comparing it with LATE in AD (LATE-AD), LATE with mixed pathology of LBD and AD (LATE-LBD + AD), and LATE alone (Pure LATE). We analyzed four cohorts of autopsy-confirmed LBD (n = 313), AD (n = 282), LBD + AD (n = 355), and aging (n = 111). We assessed the association of LATE with patient profiles including LBD subtype and AD neuropathologic change (ADNC). We studied the morphological and distributional differences between LATE-LBD and LATE-AD. By frequency analysis, we staged LATE-LBD and examined the association with cognitive impairment and genetic risk factors. Demographic analysis showed LATE associated with age in all four cohorts and the frequency of LATE was the highest in LBD + AD followed by AD, LBD, and Aging. LBD subtype and ADNC associated with LATE in LBD or AD but not in LBD + AD. Pathological analysis revealed that the hippocampal distribution of LATE was different between LATE-LBD and LATE-AD: neuronal cytoplasmic inclusions were more frequent in cornu ammonis 3 (CA3) in LATE-LBD compared to LATE-AD and abundant fine neurites composed of C-terminal truncated TDP-43 were found mainly in CA2 to subiculum in LATE-LBD, which were not as numerous in LATE-AD. Some of these fine neurites colocalized with phosphorylated α-synuclein. LATE-LBD staging showed LATE neuropathological changes spread in the dentate gyrus and brainstem earlier than in LATE-AD. The presence and prevalence of LATE in LBD associated with cognitive impairment independent of either LBD subtype or ADNC; LATE-LBD stage also associated with the genetic risk variants of TMEM106B rs1990622 and GRN rs5848. These data highlight clinicopathological and genetic features of LATE-LBD., (© 2021. The Author(s), under exclusive licence to Springer-Verlag GmbH Germany, part of Springer Nature.)
- Published
- 2022
- Full Text
- View/download PDF
16. Effects of microglial depletion and TREM2 deficiency on Aβ plaque burden and neuritic plaque tau pathology in 5XFAD mice.
- Author
-
Delizannis AT, Nonneman A, Tsering W, De Bondt A, Van den Wyngaert I, Zhang B, Meymand E, Olufemi MF, Koivula P, Maimaiti S, Trojanowski JQ, Lee VM, and Brunden KR
- Subjects
- Animals, Female, Male, Membrane Glycoproteins genetics, Mice, Mice, Knockout, Mice, Transgenic, Plaque, Amyloid chemically induced, Plaque, Amyloid genetics, Receptors, Immunologic genetics, tau Proteins administration & dosage, Membrane Glycoproteins deficiency, Microglia metabolism, Plaque, Amyloid metabolism, Receptors, Immunologic deficiency, tau Proteins toxicity
- Abstract
Dystrophic neuronal processes harboring neuritic plaque (NP) tau pathology are found in association with Aβ plaques in Alzheimer's disease (AD) brain. Microglia are also in proximity to these plaques and microglial gene variants are known risk factors in AD, including loss-of-function variants of TREM2. We have further investigated the role of Aβ plaque-associated microglia in 5XFAD mice in which NP tau pathology forms after intracerebral injection of AD brain-derived pathologic tau (AD-tau), focusing on the consequences of reduced TREM2 expression and microglial depletion after treatment with the colony-stimulating factor 1 (CSFR1) inhibitor, PLX3397. Young 5XFAD mice treated with PLX3397 had a large reduction of brain microglia, including cortical plaque-associated microglia, with a significant reduction of Aβ plaque burden in the cortex. A corresponding decrease in cortical APP-positive dystrophic processes and NP tau pathology were observed after intracerebral AD-tau injection in the PLX3397-treated 5XFAD mice. Consistent with prior reports, 5XFAD × TREM2
-/- mice showed a significant reduction of plaque-associated microglial, whereas 5XFAD × TREM2+/- mice had significantly more plaque-associated microglia than 5XFAD × TREM2-/- mice. Nonetheless, AD-tau injected 5XFAD × TREM2+/- mice showed greatly increased AT8-positive NP tau relative to 5XFAD × TREM2+/+ mice. Expression profiling revealed that 5XFAD × TREM2+/- mice had a disease-associated microglial (DAM) gene expression profile in the brain that was generally intermediate between 5XFAD × TREM2+/+ and 5XFAD × TREM2-/- mice. Microarray analysis revealed significant differences in cortical and hippocampal gene expression between AD-tau injected 5XFAD × TREM2+/- and 5XFAD × TREM2-/- mice, including pathways linked to microglial function. These data suggest there is not a simple correlation between the extent of microglia plaque interaction and plaque-associated neuritic damage. Moreover, the differences in gene expression and microglial phenotype between TREM2+/- and TREM2-/- mice suggest that the former may better model the single copy TREM2 variants associated with AD risk., (© 2021. The Author(s).)- Published
- 2021
- Full Text
- View/download PDF
17. Congeners Derived from Microtubule-Active Phenylpyrimidines Produce a Potent and Long-Lasting Paralysis of Schistosoma mansoni In Vitro.
- Author
-
Monti L, Cornec AS, Oukoloff K, Kovalevich J, Prijs K, Alle T, Brunden KR, Smith AB 3rd, El-Sakkary N, Liu LJ, Syed A, Skinner DE, Ballatore C, and Caffrey CR
- Subjects
- Animals, Humans, Paralysis, Structure-Activity Relationship, Microtubules, Schistosoma mansoni
- Abstract
Schistosomiasis is a parasitic disease that affects approximately 200 million people in developing countries. Current treatment relies on just one partially effective drug, and new drugs are needed. Tubulin and microtubules (MTs) are essential constituents of the cytoskeleton in all eukaryotic cells and considered potential drug targets to treat parasitic infections. The α- and β-tubulin of Schistosoma mansoni have ∼96% and ∼91% sequence identity to their respective human tubulins, suggesting that compounds which bind mammalian tubulin may interfere with MT-mediated functions in the parasite. To explore the potential of different classes of tubulin-binding molecules as antischistosomal leads, we completed a series of in vitro whole-organism screens of a target-based compound library against S. mansoni adults and somules (postinfective larvae), and identified multiple biologically active compounds, among which phenylpyrimidines were the most promising. Further structure-activity relationship studies of these hits identified a series of thiophen-2-yl-pyrimidine congeners, which induce a potent and long-lasting paralysis of the parasite. Moreover, compared to the originating compounds, which showed cytotoxicity values in the low nanomolar range, these new derivatives were 1-4 orders of magnitude less cytotoxic and exhibited weak or undetectable activity against mammalian MTs in a cell-based assay of MT stabilization. Given their selective antischistosomal activity and relatively simple drug-like structures, these molecules hold promise as candidates for the development of new treatments for schistosomiasis.
- Published
- 2021
- Full Text
- View/download PDF
18. In vitro amplification of pathogenic tau conserves disease-specific bioactive characteristics.
- Author
-
Xu H, O'Reilly M, Gibbons GS, Changolkar L, McBride JD, Riddle DM, Zhang B, Stieber A, Nirschl J, Kim SJ, Hoxha KH, Brunden KR, Schellenberg GD, Trojanowski JQ, and Lee VM
- Subjects
- Alzheimer Disease pathology, Animals, Brain pathology, Cells, Cultured, Conserved Sequence, Gene Amplification, Immunohistochemistry, Mice, Mice, Inbred C57BL, Neurodegenerative Diseases pathology, Neurofibrillary Tangles pathology, Primary Cell Culture, Supranuclear Palsy, Progressive pathology, Tauopathies pathology, tau Proteins genetics
- Abstract
The microtubule-associated protein tau (tau) forms hyperphosphorylated aggregates in the brains of tauopathy patients that can be pathologically and biochemically defined as distinct tau strains. Recent studies show that these tau strains exhibit strain-specific biological activities, also referred to as pathogenicities, in the tau spreading models. Currently, the specific pathogenicity of human-derived tau strains cannot be fully recapitulated by synthetic tau preformed fibrils (pffs), which are generated from recombinant tau protein. Reproducing disease-relevant tau pathology in cell and animal models necessitates the use of human brain-derived tau seeds. However, the availability of human-derived tau is extremely limited. Generation of tau variants that can mimic the pathogenicity of human-derived tau seeds would significantly extend the scale of experimental design within the field of tauopathy research. Previous studies have demonstrated that in vitro seeding reactions can amplify the beta-sheet structure of tau protein from a minute quantity of human-derived tau. However, whether the strain-specific pathogenicities of the original, human-derived tau seeds are conserved in the amplified tau strains has yet to be experimentally validated. Here, we used biochemically enriched brain-derived tau seeds from Alzheimer's disease (AD), corticobasal degeneration (CBD) and progressive supranuclear palsy (PSP) patient brains with a modified seeding protocol to template the recruitment of recombinant 2N4R (T40) tau in vitro. We quantitatively interrogated efficacy of the amplification reactions and the pathogenic fidelity of the amplified material to the original tau seeds using recently developed sporadic tau spreading models. Our data suggest that different tau strains can be faithfully amplified in vitro from tau isolated from different tauopathy brains and that the amplified tau variants retain their strain-dependent pathogenic characteristics.
- Published
- 2021
- Full Text
- View/download PDF
19. Evaluation of the Structure-Activity Relationship of Microtubule-Targeting 1,2,4-Triazolo[1,5- a ]pyrimidines Identifies New Candidates for Neurodegenerative Tauopathies.
- Author
-
Oukoloff K, Nzou G, Varricchio C, Lucero B, Alle T, Kovalevich J, Monti L, Cornec AS, Yao Y, James MJ, Trojanowski JQ, Lee VM, Smith AB 3rd, Brancale A, Brunden KR, and Ballatore C
- Subjects
- Animals, Brain metabolism, Cell Line, Cells, Cultured, Computer Simulation, Humans, Mice, Mice, Transgenic, Models, Molecular, Molecular Docking Simulation, Neurons drug effects, Rats, Structure-Activity Relationship, Microtubules drug effects, Neurodegenerative Diseases drug therapy, Pyrimidines chemistry, Pyrimidines pharmacology, Tauopathies drug therapy, Triazoles chemistry, Triazoles pharmacology
- Abstract
Studies in tau and Aβ plaque transgenic mouse models demonstrated that brain-penetrant microtubule (MT)-stabilizing compounds, including the 1,2,4-triazolo[1,5- a ]pyrimidines, hold promise as candidate treatments for Alzheimer's disease and related neurodegenerative tauopathies. Triazolopyrimidines have already been investigated as anticancer agents; however, the antimitotic activity of these compounds does not always correlate with stabilization of MTs in cells. Indeed, previous studies from our laboratories identified a critical role for the fragment linked at C6 in determining whether triazolopyrimidines promote MT stabilization or, conversely, disrupt MT integrity in cells. To further elucidate the structure-activity relationship (SAR) and to identify potentially improved MT-stabilizing candidates for neurodegenerative disease, a comprehensive set of 68 triazolopyrimidine congeners bearing structural modifications at C6 and/or C7 was designed, synthesized, and evaluated. These studies expand upon prior understanding of triazolopyrimidine SAR and enabled the identification of novel analogues that, relative to the existing lead, exhibit improved physicochemical properties, MT-stabilizing activity, and pharmacokinetics.
- Published
- 2021
- Full Text
- View/download PDF
20. Characterization of tau binding by gosuranemab.
- Author
-
Sopko R, Golonzhka O, Arndt J, Quan C, Czerkowicz J, Cameron A, Smith B, Murugesan Y, Gibbons G, Kim SJ, Trojanowski JQ, Lee VMY, Brunden KR, Graham DL, Weinreb PH, and Hering H
- Subjects
- Animals, Basal Ganglia Diseases metabolism, Mice, Transgenic, Neurons metabolism, Supranuclear Palsy, Progressive metabolism, Tauopathies metabolism, Tauopathies pathology, Alzheimer Disease metabolism, Antibodies, Monoclonal, Humanized metabolism, Brain metabolism, tau Proteins metabolism
- Abstract
Deposition of tau aggregates in the brain is a pathological hallmark of several neurodegenerative diseases, termed tauopathies, such as Alzheimer's disease (AD), corticobasal degeneration, and progressive supranuclear palsy (PSP). As transcellular spread of pathological tau aggregates has been implicated in disease progression, immunotherapy is being considered as a treatment for tauopathies. Here we report a detailed biochemical and biophysical characterization of the tau-binding properties of gosuranemab, a humanized monoclonal antibody directed against N-terminal tau that is currently being investigated as a treatment for AD. Binding experiments showed that gosuranemab exhibited high affinity for tau monomer, tau fibrils, and insoluble tau from different tauopathies. Epitope mapping studies conducted using X-ray crystallography and mutagenesis showed that gosuranemab bound to human tau residues 15-22. Immunodepletion of pathological human brain homogenates and transgenic mouse interstitial fluid (ISF) with gosuranemab resulted in reduced tau aggregation in tau biosensor cells. Preincubation of seed-competent AD-tau with gosuranemab significantly inhibited tau aggregation in mouse primary cortical neurons. Gosuranemab also significantly reduced unbound N-terminal tau in cerebrospinal fluid (CSF) from individuals with PSP and AD, and in ISF and CSF of treated transgenic mice. These results are consistent with the >90% target engagement observed in the CSF of some clinical trial dosing cohorts and support the evaluation of gosuranemab as a potential treatment for AD., (Copyright © 2020. Published by Elsevier Inc.)
- Published
- 2020
- Full Text
- View/download PDF
21. Conformation-selective tau monoclonal antibodies inhibit tau pathology in primary neurons and a mouse model of Alzheimer's disease.
- Author
-
Gibbons GS, Kim SJ, Wu Q, Riddle DM, Leight SN, Changolkar L, Xu H, Meymand ES, O'Reilly M, Zhang B, Brunden KR, Trojanowski JQ, and Lee VMY
- Subjects
- Animals, Disease Models, Animal, Humans, Mice, Neurons drug effects, tau Proteins drug effects, Alzheimer Disease pathology, Antibodies, Monoclonal pharmacology, Neurons pathology, tau Proteins antagonists & inhibitors
- Abstract
Background: The spread of tau pathology in Alzheimer's disease (AD) is mediated by cell-to-cell transmission of pathological tau seeds released from neurons that, upon internalization by recipient neurons, template the misfolding of naïve cellular tau, thereby propagating fibrillization. We hypothesize that anti-tau monoclonal antibodies (mAbs) that selectively bind to pathological tau seeds will inhibit propagation of tau aggregates and reduce the spread of tau pathology in vivo., Methods: We inoculated mice with human AD brain-derived extracts containing tau paired helical filaments (AD-tau) and identified two novel mAbs, DMR7 and SKT82, that selectively bind to a misfolded pathological conformation of tau relative to recombinant tau monomer. To evaluate the effects of these mAbs on the spread of pathological tau in vivo, 5xFAD mice harboring significant brain Aβ plaque burden were unilaterally injected with AD-tau in the hippocampus, to initiate the formation of neuritic plaque (NP) tau pathology, and were treated weekly with intraperitoneal (i.p.) injections of DMR7, SKT82, or IgG isotype control mAbs., Results: DMR7 and SKT82 bind epitopes comprised of the proline-rich domain and c-terminal region of tau and binding is reduced upon disruption of the pathological conformation of AD-tau by chemical and thermal denaturation. We found that both DMR7 and SKT82 immunoprecipitate pathological tau and significantly reduce the seeding of cellular tau aggregates induced by AD-tau in primary neurons by 60.5 + 13.8% and 82.2 + 8.3%, respectively, compared to IgG control. To investigate the mechanism of mAb inhibition, we generated pH-sensitive fluorophore-labeled recombinant tau fibrils seeded by AD-tau to track internalization of tau seeds and demonstrate that the conformation-selective tau mAbs inhibit the internalization of tau seeds. DMR7 and SKT82 treatment reduced hyperphosphorylated NP tau as measured with AT8 immunohistochemistry (IHC) staining, but did not achieve statistical significance in the contralateral cortex and SKT82 significantly reduced tau pathology in the ipsilateral hippocampus by 24.2%; p = 0.044., Conclusions: These findings demonstrate that conformation-selective tau mAbs, DMR7 and SKT82, inhibit tau pathology in primary neurons by preventing the uptake of tau seeds and reduce tau pathology in vivo, providing potential novel therapeutic candidates for the treatment of AD.
- Published
- 2020
- Full Text
- View/download PDF
22. Correction of microtubule defects within Aβ plaque-associated dystrophic axons results in lowered Aβ release and plaque deposition.
- Author
-
Yao Y, Nzou G, Alle T, Tsering W, Maimaiti S, Trojanowski JQ, Lee VM, Ballatore C, and Brunden KR
- Subjects
- Amyloid beta-Peptides metabolism, Animals, Brain metabolism, Brain pathology, Disease Models, Animal, Female, Humans, Male, Mice, Mice, Transgenic, Microtubules pathology, Axons pathology, Brain drug effects, Hydrocarbons, Halogenated pharmacology, Microtubules drug effects, Plaque, Amyloid pathology, Triazoles pharmacology
- Abstract
The hallmark pathologies of the Alzheimer's disease (AD) brain are amyloid beta (Aβ)-containing senile plaques and neurofibrillary tangles formed from the microtubule (MT)-binding tau protein. Tau becomes hyperphosphorylated and disengages from MTs in AD, with evidence of resulting MT structure/function defects. Brain-penetrant MT-stabilizing compounds can normalize MTs and axonal transport in mouse models with tau pathology, thereby reducing neuron loss and decreasing tau pathology. MT dysfunction is also observed in dystrophic axons adjacent to Aβ plaques, resulting in accumulation of amyloid precursor protein (APP) and BACE1 with the potential for enhanced localized Aβ generation. We have examined whether the brain-penetrant MT-stabilizing compound CNDR-51657 might decrease plaque-associated axonal dystrophy and Aβ release in 5XFAD mice that develop an abundance of Aβ plaques. Administration of CNDR-51657 to 1.5-month-old male and female 5XFAD mice for 4 or 7 weeks led to decreased soluble brain Aβ that coincided with reduced APP and BACE1 levels, resulting in decreased formation of insoluble Aβ deposits. These data suggest a vicious cycle whereby initial Aβ plaque formation causes MT disruption in nearby axons, resulting in the local accumulation of APP and BACE1 that facilitates additional Aβ generation and plaque deposition. The ability of a MT-stabilizing compound to attenuate this cycle, and also reduce deficits resulting from reduced tau binding to MTs, suggests that molecules of this type hold promise as potential AD therapeutics., (© 2020 the Alzheimer's Association.)
- Published
- 2020
- Full Text
- View/download PDF
23. Compound screening in cell-based models of tau inclusion formation: Comparison of primary neuron and HEK293 cell assays.
- Author
-
Crowe A, Henderson MJ, Anderson J, Titus SA, Zakharov A, Simeonov A, Buist A, Delay C, Moechars D, Trojanowski JQ, Lee VM, and Brunden KR
- Subjects
- Alzheimer Disease metabolism, Alzheimer Disease pathology, Animals, Dopamine D2 Receptor Antagonists chemistry, Dopamine D2 Receptor Antagonists metabolism, Dopamine D2 Receptor Antagonists pharmacology, HEK293 Cells, Humans, Mice, Microscopy, Fluorescence, Neurons cytology, Neurons metabolism, Rats, Small Molecule Libraries chemistry, Small Molecule Libraries metabolism, tau Proteins antagonists & inhibitors, tau Proteins genetics, Protein Aggregates drug effects, Small Molecule Libraries pharmacology, tau Proteins metabolism
- Abstract
The hallmark pathological features of Alzheimer's disease (AD) brains are senile plaques, comprising β-amyloid (Aβ) peptides, and neuronal inclusions formed from tau protein. These plaques form 10-20 years before AD symptom onset, whereas robust tau pathology is more closely associated with symptoms and correlates with cognitive status. This temporal sequence of AD pathology development, coupled with repeated clinical failures of Aβ-directed drugs, suggests that molecules that reduce tau inclusions have therapeutic potential. Few tau-directed drugs are presently in clinical testing, in part because of the difficulty in identifying molecules that reduce tau inclusions. We describe here two cell-based assays of tau inclusion formation that we employed to screen for compounds that inhibit tau pathology: a HEK293 cell-based tau overexpression assay, and a primary rat cortical neuron assay with physiological tau expression. Screening a collection of ∼3500 pharmaceutical compounds with the HEK293 cell tau aggregation assay, we obtained only a low number of hit compounds. Moreover, these compounds generally failed to inhibit tau inclusion formation in the cortical neuron assay. We then screened the Prestwick library of mostly approved drugs in the cortical neuron assay, leading to the identification of a greater number of tau inclusion inhibitors. These included four dopamine D2 receptor antagonists, with D2 receptors having previously been suggested to regulate tau inclusions in a Caenorhabditis elegans model. These results suggest that neurons, the cells most affected by tau pathology in AD, are very suitable for screening for tau inclusion inhibitors.
- Published
- 2020
- Full Text
- View/download PDF
24. Characterization of novel conformation-selective α-synuclein antibodies as potential immunotherapeutic agents for Parkinson's disease.
- Author
-
Henderson MX, Covell DJ, Chung CH, Pitkin RM, Sandler RM, Decker SC, Riddle DM, Zhang B, Gathagan RJ, James MJ, Trojanowski JQ, Brunden KR, Lee VMY, and Luk KC
- Subjects
- Animals, Antibodies, Monoclonal chemistry, Antibodies, Monoclonal genetics, Cells, Cultured, Dose-Response Relationship, Immunologic, Female, Humans, Lewy Body Disease genetics, Male, Mice, Mice, Inbred BALB C, Parkinson Disease genetics, alpha-Synuclein chemistry, alpha-Synuclein genetics, Antibodies, Monoclonal administration & dosage, Immunotherapy methods, Lewy Body Disease immunology, Lewy Body Disease therapy, Parkinson Disease immunology, Parkinson Disease therapy, alpha-Synuclein administration & dosage
- Abstract
Parkinson's disease (PD) and dementia with Lewy bodies (DLB) are progressive neurodegenerative diseases for which there is no disease-modifying treatment. PD and DLB are characterized by aggregation of the synaptic protein α-synuclein, and there is compelling evidence to suggest that progression of these diseases is associated with the trans-cellular spread of pathogenic α-synuclein through the brains of afflicted individuals. Therapies targeting extracellular, pathogenic α-synuclein may therefore hold promise for slowing or halting disease progression. In this regard, it has been suggested that highly-selective antibodies can be administered as therapeutic agents targeting pathogenic proteins. In the current study, we screened a series of antibodies using multiple selection criterion to identify those that selectively bind pathogenic α-synuclein and show potent inhibition of pathology seeding in a neuronal model of α-synucleinopathy. A lead antibody was tested in a mouse model of PD, and it was able to reduce the spread of α-synuclein pathology in the brain and attenuate dopamine reductions in the striatum. This study highlights the therapeutic potential of α-synuclein immunotherapy for the treatment of PD and DLB, and provides a framework for screening of α-synuclein antibodies to identify those with preferred properties., Competing Interests: Declaration of Competing Interest This research was partially funded by GlaxoSmithKline., (Copyright © 2019. Published by Elsevier Inc.)
- Published
- 2020
- Full Text
- View/download PDF
25. 1,2,4-Triazolo[1,5-a]pyrimidines in drug design.
- Author
-
Oukoloff K, Lucero B, Francisco KR, Brunden KR, and Ballatore C
- Subjects
- Animals, Drug Design, Humans, Neoplasms drug therapy, Parasitic Diseases drug therapy, Pyrimidines therapeutic use, Structure-Activity Relationship, Triazoles therapeutic use, Chemistry, Pharmaceutical methods, Pyrimidines chemistry, Triazoles chemistry
- Abstract
The 1,2,4-triazolo[1,5-a]pyrimidine (TP) heterocycle, in spite of its relatively simple structure, has proved to be remarkably versatile as evidenced by its use in many different applications reported over the years in different areas of drug design. For example, as the ring system of TPs is isoelectronic with that of purines, this heterocycle has been proposed as a possible surrogate of the purine ring. However, depending on the choice of substituents, the TP ring has also been described as a potentially viable bio-isostere of the carboxylic acid functional group and of the N-acetyl fragment of ε-N-acetylated lysine. In addition, the metal-chelating properties of the TP ring have also been exploited to generate candidate treatments for cancer and parasitic diseases. In the present review article, we discuss recent applications of the TP scaffold in medicinal chemistry, and provide an overview of its properties and methods of synthesis., (Copyright © 2019 Elsevier Masson SAS. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
26. A brain-penetrant triazolopyrimidine enhances microtubule-stability, reduces axonal dysfunction and decreases tau pathology in a mouse tauopathy model.
- Author
-
Zhang B, Yao Y, Cornec AS, Oukoloff K, James MJ, Koivula P, Trojanowski JQ, Smith AB 3rd, Lee VM, Ballatore C, and Brunden KR
- Subjects
- Alzheimer Disease drug therapy, Alzheimer Disease metabolism, Animals, Axonal Transport drug effects, Axons drug effects, Axons metabolism, Brain metabolism, Disease Models, Animal, Humans, Mice, Microtubules metabolism, Neurons drug effects, Neurons metabolism, Tauopathies metabolism, tau Proteins metabolism, Brain drug effects, Microtubules drug effects, Quinazolines pharmacology, Tauopathies drug therapy, Triazoles pharmacology, tau Proteins drug effects
- Abstract
Background: Alzheimer's disease (AD) and related tauopathies are neurodegenerative diseases that are characterized by the presence of insoluble inclusions of the protein tau within brain neurons and often glia. Tau is normally found associated with axonal microtubules (MTs) in the brain, and in tauopathies this MT binding is diminished due to tau hyperphosphorylation. As MTs play a critical role in the movement of cellular constituents within neurons via axonal transport, it is likely that the dissociation of tau from MTs alters MT structure and axonal transport, and there is evidence of this in tauopathy mouse models as well as in AD brain. We previously demonstrated that different natural products which stabilize MTs by interacting with β-tubulin at the taxane binding site provide significant benefit in transgenic mouse models of tauopathy. More recently, we have reported on a series of MT-stabilizing triazolopyrimidines (TPDs), which interact with β-tubulin at the vinblastine binding site, that exhibit favorable properties including brain penetration and oral bioavailability. Here, we have examined a prototype TPD example, CNDR-51657, in a secondary prevention study utilizing aged tau transgenic mice., Methods: 9-Month old female PS19 mice with a low amount of existing tau pathology received twice-weekly administration of vehicle, or 3 or 10 mg/kg of CNDR-51657, for 3 months. Mice were examined in the Barnes maze at the end of the dosing period, and brain tissue and optic nerves were examined immunohistochemically or biochemically for changes in MT density, axonal dystrophy, and tau pathology. Mice were also assessed for changes in organ weights and blood cell numbers., Results: CNDR-51657 caused a significant amelioration of the MT deficit and axonal dystrophy observed in vehicle-treated aged PS19 mice. Moreover, PS19 mice receiving CNDR-51657 had significantly lower tau pathology, with a trend toward improved Barnes maze performance. Importantly, no adverse effects were observed in the compound-treated mice, including no change in white blood cell counts as is often observed in cancer patients receiving high doses of MT-stabilizing drugs., Conclusions: A brain-penetrant MT-stabilizing TPD can safely correct MT and axonal deficits in an established mouse model of tauopathy, resulting in reduced tau pathology.
- Published
- 2018
- Full Text
- View/download PDF
27. Brain-Penetrant Triazolopyrimidine and Phenylpyrimidine Microtubule Stabilizers as Potential Leads to Treat Human African Trypanosomiasis.
- Author
-
Monti L, Wang SC, Oukoloff K, Smith AB 3rd, Brunden KR, Caffrey CR, and Ballatore C
- Subjects
- Dose-Response Relationship, Drug, Humans, Microtubules chemistry, Molecular Structure, Parasitic Sensitivity Tests, Pyrimidines chemistry, Pyrimidines metabolism, Structure-Activity Relationship, Trypanocidal Agents chemistry, Trypanocidal Agents metabolism, Brain metabolism, Microtubules metabolism, Pyrimidines pharmacology, Trypanocidal Agents pharmacology, Trypanosoma brucei brucei drug effects, Trypanosomiasis, African drug therapy
- Abstract
In vitro whole-organism screens of Trypanosoma brucei with representative examples of brain-penetrant microtubule (MT)-stabilizing agents identified lethal triazolopyrimidines and phenylpyrimidines with sub-micromolar potency. In mammalian cells, these antiproliferative compounds disrupt MT integrity and decrease total tubulin levels. Their parasiticidal potency, combined with their generally favorable pharmacokinetic properties, which include oral bioavailability and brain penetration, suggest that these compounds are potential leads against human African trypanosomiasis., (© 2018 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2018
- Full Text
- View/download PDF
28. Design, synthesis and evaluation of photoactivatable derivatives of microtubule (MT)-active [1,2,4]triazolo[1,5-a]pyrimidines.
- Author
-
Oukoloff K, Kovalevich J, Cornec AS, Yao Y, Owyang ZA, James M, Trojanowski JQ, Lee VM, Smith AB 3rd, Brunden KR, and Ballatore C
- Subjects
- Fluorescent Dyes chemistry, HEK293 Cells, Humans, Molecular Structure, Pyrimidines chemical synthesis, Pyrimidines chemistry, Triazoles chemical synthesis, Triazoles chemistry, Drug Design, Fluorescent Dyes chemical synthesis, Microtubules chemistry, Pyrimidines pharmacology, Triazoles pharmacology
- Abstract
The [1,2,4]triazolo[1,5-a]pyrimidines comprise a promising class of non-naturally occurring microtubule (MT)-active compounds. Prior studies revealed that different triazolopyrimidine substitutions can yield molecules that either promote MT stabilization or disrupt MT integrity. These differences can have important ramifications in the therapeutic applications of triazolopyrimidines and suggest that different analogues may exhibit different binding modes within the same site or possibly interact with tubulin/MTs at alternative binding sites. To help discern these possibilities, a series of photoactivatable triazolopyrimidine congeners was designed, synthesized and evaluated in cellular assays with the goal of identifying candidate probes for photoaffinity labeling experiments. These studies led to the identification of different derivatives that incorporate a diazirine ring in the amine substituent at position 7 of the triazolopyrimidine heterocycle, resulting in molecules that either promote stabilization of MTs or disrupt MT integrity. These photoactivatable candidate probes hold promise to investigate the mode of action of MT-active triazolopyrimidines., (Copyright © 2018 Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
29. Amyloid-β plaques enhance Alzheimer's brain tau-seeded pathologies by facilitating neuritic plaque tau aggregation.
- Author
-
He Z, Guo JL, McBride JD, Narasimhan S, Kim H, Changolkar L, Zhang B, Gathagan RJ, Yue C, Dengler C, Stieber A, Nitla M, Coulter DA, Abel T, Brunden KR, Trojanowski JQ, and Lee VM
- Subjects
- Alzheimer Disease metabolism, Animals, Axons metabolism, Hippocampus metabolism, Humans, Mice, Neurofibrillary Tangles, Alzheimer Disease pathology, Amyloid beta-Peptides metabolism, Neurites metabolism, tau Proteins metabolism
- Abstract
Alzheimer's disease (AD) is characterized by extracellular amyloid-β (Aβ) plaques and intracellular tau inclusions. However, the exact mechanistic link between these two AD lesions remains enigmatic. Through injection of human AD-brain-derived pathological tau (AD-tau) into Aβ plaque-bearing mouse models that do not overexpress tau, we recapitulated the formation of three major types of AD-relevant tau pathologies: tau aggregates in dystrophic neurites surrounding Aβ plaques (NP tau), AD-like neurofibrillary tangles (NFTs) and neuropil threads (NTs). These distinct tau pathologies have different temporal onsets and functional consequences on neural activity and behavior. Notably, we found that Aβ plaques created a unique environment that facilitated the rapid amplification of proteopathic AD-tau seeds into large tau aggregates, initially appearing as NP tau, which was followed by the formation and spread of NFTs and NTs, likely through secondary seeding events. Our study provides insights into a new multistep mechanism underlying Aβ plaque-associated tau pathogenesis.
- Published
- 2018
- Full Text
- View/download PDF
30. Altered microtubule dynamics in neurodegenerative disease: Therapeutic potential of microtubule-stabilizing drugs.
- Author
-
Brunden KR, Lee VM, Smith AB 3rd, Trojanowski JQ, and Ballatore C
- Subjects
- Animals, Humans, Microtubules drug effects, Tubulin Modulators chemistry, Tubulin Modulators pharmacology, Microtubules metabolism, Neurodegenerative Diseases drug therapy, Neurodegenerative Diseases metabolism, Tubulin Modulators therapeutic use
- Abstract
Many neurodegenerative diseases are characterized by deficiencies in neuronal axonal transport, a process in which cellular cargo is shuttled with the aid of molecular motors from the cell body to axonal termini and back along microtubules (MTs). Proper axonal transport is critical to the normal functioning of neurons, and impairments in this process could contribute to the neuronal damage and death that is characteristic of neurodegenerative disease. Although the causes of axonal transport abnormalities may vary among the various neurodegenerative conditions, in many cases it appears that the transport deficiencies result from a diminution of axonal MT stability. Here we review the evidence of MT abnormalities in a number of neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis and traumatic brain injury, and highlight the potential benefit of MT-stabilizing agents in improving axonal transport and nerve function in these diseases. Moreover, we discuss the challenges associated with the utilization of MT-stabilizing drugs as therapeutic candidates for neurodegenerative conditions., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
31. Evaluation of Oxetan-3-ol, Thietan-3-ol, and Derivatives Thereof as Bioisosteres of the Carboxylic Acid Functional Group.
- Author
-
Lassalas P, Oukoloff K, Makani V, James M, Tran V, Yao Y, Huang L, Vijayendran K, Monti L, Trojanowski JQ, Lee VM, Kozlowski MC, Smith AB 3rd, Brunden KR, and Ballatore C
- Abstract
The oxetane ring serves as an isostere of the carbonyl moiety, suggesting that oxetan-3-ol may be considered as a potential surrogate of the carboxylic acid functional group. To investigate this structural unit, as well as thietan-3-ol and the corresponding sulfoxide and sulfone derivatives, as potential carboxylic acid bioisosteres, a set of model compounds has been designed, synthesized, and evaluated for physicochemical properties. Similar derivatives of the cyclooxygenase inhibitor, ibuprofen, were also synthesized and evaluated for inhibition of eicosanoid biosynthesis in vitro. Collectively, the data suggest that oxetan-3-ol, thietan-3-ol, and related structures hold promise as isosteric replacements of the carboxylic acid moiety.
- Published
- 2017
- Full Text
- View/download PDF
32. Multitargeted Imidazoles: Potential Therapeutic Leads for Alzheimer's and Other Neurodegenerative Diseases.
- Author
-
Cornec AS, Monti L, Kovalevich J, Makani V, James MJ, Vijayendran KG, Oukoloff K, Yao Y, Lee VM, Trojanowski JQ, Smith AB 3rd, Brunden KR, and Ballatore C
- Subjects
- Alzheimer Disease drug therapy, Animals, Arachidonate 5-Lipoxygenase metabolism, Chemistry Techniques, Synthetic, Cyclooxygenase Inhibitors chemistry, Drug Evaluation, Preclinical methods, Female, Humans, Imidazoles chemistry, Leukotrienes biosynthesis, Lipoxygenase Inhibitors chemistry, Male, Mice, Inbred Strains, Microtubules drug effects, Microtubules metabolism, Molecular Targeted Therapy, Prostaglandins metabolism, Rats, Cyclooxygenase Inhibitors pharmacology, Imidazoles pharmacology, Lipoxygenase Inhibitors pharmacology, Neurodegenerative Diseases drug therapy, Structure-Activity Relationship
- Abstract
Alzheimer's disease (AD) is a complex, multifactorial disease in which different neuropathological mechanisms are likely involved, including those associated with pathological tau and Aβ species as well as neuroinflammation. In this context, the development of single multitargeted therapeutics directed against two or more disease mechanisms could be advantageous. Starting from a series of 1,5-diarylimidazoles with microtubule (MT)-stabilizing activity and structural similarities with known NSAIDs, we conducted structure-activity relationship studies that led to the identification of multitargeted prototypes with activities as MT-stabilizing agents and/or inhibitors of the cyclooxygenase (COX) and 5-lipoxygenase (5-LOX) pathways. Several examples are brain-penetrant and exhibit balanced multitargeted in vitro activity in the low μM range. As brain-penetrant MT-stabilizing agents have proven effective against tau-mediated neurodegeneration in animal models, and because COX- and 5-LOX-derived eicosanoids are thought to contribute to Aβ plaque deposition, these 1,5-diarylimidazoles provide tools to explore novel multitargeted strategies for AD and other neurodegenerative diseases.
- Published
- 2017
- Full Text
- View/download PDF
33. Non-Naturally Occurring Small Molecule Microtubule-Stabilizing Agents: A Potential Tactic for CNS-Directed Therapies.
- Author
-
Ballatore C, Brunden KR, Trojanowski JQ, Lee VM, and Smith AB 3rd
- Subjects
- Alzheimer Disease drug therapy, Alzheimer Disease metabolism, Animals, Biological Products administration & dosage, Central Nervous System drug effects, Central Nervous System metabolism, Drug Delivery Systems trends, Humans, Microtubules drug effects, Biological Products chemistry, Biological Products metabolism, Microtubules metabolism, Neurodegenerative Diseases drug therapy, Neurodegenerative Diseases metabolism
- Abstract
Several independent studies indicate that microtubule (MT)-stabilizing agents hold considerable promise as candidate therapeutics for a wide spectrum of conditions of the central nervous system (CNS), from brain tumors to spinal cord injury, as well as a number of neurodegenerative diseases, including Alzheimer's disease, frontotemporal lobar degeneration, Parkinson's disease, and amyotrophic lateral sclerosis. Although the identification and development of candidate compounds for CNS-directed MT-stabilizing therapies has been a challenge in drug discovery for many years, a growing number of molecules have now been identified that exhibit both MT-stabilizing activity and brain penetration. In this Viewpoint, we will highlight the potential utility of MT-active triazolopyrimidines, phenylpyrimidines, and related classes of non-naturally occurring small molecules that exhibit favorable druglike properties, including brain penetration and oral bioavailability. The mode of action of these small molecules has not as yet been fully elucidated at the molecular level. However, based on all available data, compounds from these classes appear to act on MTs in a potentially unique manner. Further characterization of these molecules may have important ramifications for drug discovery, especially in the area of CNS diseases., Competing Interests: The authors declare no competing financial interest.
- Published
- 2017
- Full Text
- View/download PDF
34. Therapeutic strategies for the treatment of tauopathies: Hopes and challenges.
- Author
-
Khanna MR, Kovalevich J, Lee VM, Trojanowski JQ, and Brunden KR
- Subjects
- Alzheimer Disease pathology, Brain pathology, Frontotemporal Lobar Degeneration pathology, Humans, tau Proteins metabolism, Alzheimer Disease drug therapy, Frontotemporal Lobar Degeneration drug therapy, Tauopathies drug therapy
- Abstract
A group of neurodegenerative diseases referred to as tauopathies are characterized by the presence of brain cells harboring inclusions of pathological species of the tau protein. These disorders include Alzheimer's disease and frontotemporal lobar degeneration due to tau pathology, including progressive supranuclear palsy, corticobasal degeneration, and Pick's disease. Tau is normally a microtubule (MT)-associated protein that appears to play an important role in ensuring proper axonal transport, but in tauopathies tau becomes hyperphosphorylated and disengages from MTs, with consequent misfolding and deposition into inclusions that mainly affect neurons but also glia. A body of experimental evidence suggests that the development of tau inclusions leads to the neurodegeneration observed in tauopathies, and there is a growing interest in developing tau-directed therapeutic agents. The following review provides a summary of strategies under investigation for the potential treatment of tauopathies, highlighting both the promises and challenges associated with these various therapeutic approaches., (Copyright © 2016 The Alzheimer's Association. Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
35. Evaluation of the brain-penetrant microtubule-stabilizing agent, dictyostatin, in the PS19 tau transgenic mouse model of tauopathy.
- Author
-
Makani V, Zhang B, Han H, Yao Y, Lassalas P, Lou K, Paterson I, Lee VM, Trojanowski JQ, Ballatore C, Smith AB 3rd, and Brunden KR
- Abstract
Neurodegenerative disorders referred to as tauopathies, which includes Alzheimer's disease (AD), are characterized by insoluble deposits of the tau protein within neuron cell bodies and dendritic processes in the brain. Tau is normally associated with microtubules (MTs) in axons, where it provides MT stabilization and may modulate axonal transport. However, tau becomes hyperphosphorylated and dissociates from MTs in tauopathies, with evidence of reduced MT stability and defective axonal transport. This has led to the hypothesis that MT-stabilizing drugs may have potential for the treatment of tauopathies. Prior studies demonstrated that the brain-penetrant MT-stabilizing drug, epothilone D, had salutary effects in transgenic (Tg) mouse models of tauopathy, improving MT density and axonal transport, while reducing axonal dystrophy. Moreover, epothilone D enhanced cognitive performance and decreased hippocampal neuron loss, with evidence of reduced tau pathology. To date, epothilone D has been the only non-peptide small molecule MT-stabilizing agent to be evaluated in Tg tau mice. Herein, we demonstrate the efficacy of another small molecule brain-penetrant MT-stabilizing agent, dictyostatin, in the PS19 tau Tg mouse model. Although dictyostatin was poorly tolerated at once-weekly doses of 1 mg/kg or 0.3 mg/kg, likely due to gastrointestinal (GI) complications, a dictyostatin dose of 0.1 mg/kg was better tolerated, such that the majority of 6-month old PS19 mice, which harbor a moderate level of brain tau pathology, completed a 3-month dosing study without evidence of significant body weight loss. Importantly, as previously observed with epothilone D, the dictyostatin-treated PS19 mice displayed improved MT density and reduced axonal dystrophy, with a reduction of tau pathology and a trend toward increased hippocampal neuron survival relative to vehicle-treated PS19 mice. Thus, despite evidence of dose-limiting peripheral side effects, the observed positive brain outcomes in dictyostatin-treated aged PS19 mice reinforces the concept that MT-stabilizing compounds have significant potential for the treatment of tauopathies.
- Published
- 2016
- Full Text
- View/download PDF
36. The Dynamics and Turnover of Tau Aggregates in Cultured Cells: INSIGHTS INTO THERAPIES FOR TAUOPATHIES.
- Author
-
Guo JL, Buist A, Soares A, Callaerts K, Calafate S, Stevenaert F, Daniels JP, Zoll BE, Crowe A, Brunden KR, Moechars D, and Lee VM
- Subjects
- Autophagy, Cell Line, Humans, Kinetics, Lysosomes metabolism, Proteasome Endopeptidase Complex metabolism, Protein Aggregates, Proteolysis, Solubility, Tauopathies drug therapy, Ubiquitination, tau Proteins metabolism
- Abstract
Filamentous tau aggregates, the hallmark lesions of Alzheimer disease (AD), play key roles in neurodegeneration. Activation of protein degradation systems has been proposed to be a potential strategy for removing pathological tau, but it remains unclear how effectively tau aggregates can be degraded by these systems. By applying our previously established cellular model system of AD-like tau aggregate induction using preformed tau fibrils, we demonstrate that tau aggregates induced in cells with regulated expression of full-length mutant tau can be gradually cleared when soluble tau expression is suppressed. This clearance is at least partially mediated by the autophagy-lysosome pathway, although both the ubiquitin-proteasome system and the autophagy-lysosome pathway are deficient in handling large tau aggregates. Importantly, residual tau aggregates left after the clearance phase leads to a rapid reinstatement of robust tau pathology once soluble tau expression is turned on again. Moreover, we succeeded in generating monoclonal cells persistently carrying tau aggregates without obvious cytotoxicity. Live imaging of GFP-tagged tau aggregates showed that tau inclusions are dynamic structures constantly undergoing "fission" and "fusion," which facilitate stable propagation of tau pathology in dividing cells. These findings provide a greater understanding of cell-to-cell transmission of tau aggregates in dividing cells and possibly neurons., (© 2016 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
37. Characterization of Brain-Penetrant Pyrimidine-Containing Molecules with Differential Microtubule-Stabilizing Activities Developed as Potential Therapeutic Agents for Alzheimer's Disease and Related Tauopathies.
- Author
-
Kovalevich J, Cornec AS, Yao Y, James M, Crowe A, Lee VM, Trojanowski JQ, Smith AB 3rd, Ballatore C, and Brunden KR
- Subjects
- Animals, Biological Availability, Blood-Brain Barrier drug effects, Cell Division drug effects, Dose-Response Relationship, Drug, Female, Hydrocarbons, Halogenated pharmacokinetics, Male, Mice, Neurons drug effects, Proteasome Endopeptidase Complex drug effects, Pyrimidines pharmacokinetics, Triazoles pharmacokinetics, Tubulin genetics, Tubulin metabolism, tau Proteins metabolism, Alzheimer Disease drug therapy, Hydrocarbons, Halogenated therapeutic use, Microtubules drug effects, Pyrimidines pharmacology, Pyrimidines therapeutic use, Tauopathies drug therapy, Triazoles therapeutic use
- Abstract
The microtubule (MT)-stabilizing protein tau disengages from MTs and forms intracellular inclusions known as neurofibrillary tangles in Alzheimer's disease and related tauopathies. Reduced tau binding to MTs in tauopathies may contribute to neuronal dysfunction through decreased MT stabilization and disrupted axonal transport. Thus, the introduction of brain-penetrant MT-stabilizing compounds might normalize MT dynamics and axonal deficits in these disorders. We previously described a number of phenylpyrimidines and triazolopyrimidines (TPDs) that induce tubulin post-translational modifications indicative of MT stabilization. We now further characterize the biologic properties of these small molecules, and our results reveal that these compounds can be divided into two general classes based on the cellular response they evoke. One group composed of the phenylpyrimidines and several TPD examples showed a bell-shaped concentration-response effect on markers of MT stabilization in cellular assays. Moreover, these compounds induced proteasome-dependent degradation of α- and β-tubulin and caused altered MT morphology in both dividing cells and neuron cultures. In contrast, a second group comprising a subset of TPD molecules (TPD+) increased markers of stable MTs in a concentration-dependent manner in dividing cells and in neurons without affecting total tubulin levels or disrupting MT architecture. Moreover, an example TPD+ compound was shown to increase MTs in a neuron culture model with induced tau hyperphosphorylation and associated MT deficits. Several TPD+ compounds were shown to be both brain penetrant and orally bioavailable, and a TPD+ example increased MT stabilization in the mouse brain, making these compounds potential candidate therapeutics for neurodegenerative tauopathies such as Alzheimer's disease., (Copyright © 2016 by The American Society for Pharmacology and Experimental Therapeutics.)
- Published
- 2016
- Full Text
- View/download PDF
38. Structure Property Relationships of Carboxylic Acid Isosteres.
- Author
-
Lassalas P, Gay B, Lasfargeas C, James MJ, Tran V, Vijayendran KG, Brunden KR, Kozlowski MC, Thomas CJ, Smith AB 3rd, Huryn DM, and Ballatore C
- Subjects
- Chemistry, Pharmaceutical, Mass Spectrometry, Models, Molecular, Molecular Structure, Structure-Activity Relationship, Carboxylic Acids chemistry, Phenylpropionates chemistry, Plasma chemistry, Plasma drug effects
- Abstract
The replacement of a carboxylic acid with a surrogate structure, or (bio)-isostere, is a classical strategy in medicinal chemistry. The general underlying principle is that by maintaining the features of the carboxylic acid critical for biological activity, but appropriately modifying the physicochemical properties, improved analogs may result. In this context, a systematic assessment of the physicochemical properties of carboxylic acid isosteres would be desirable to enable more informed decisions of potential replacements to be used for analog design. Herein we report the structure-property relationships (SPR) of 35 phenylpropionic acid derivatives, in which the carboxylic acid moiety is replaced with a series of known isosteres. The data set generated provides an assessment of the relative impact on the physicochemical properties that these replacements may have compared to the carboxylic acid analog. As such, this study presents a framework for how to rationally apply isosteric replacements of the carboxylic acid functional group.
- Published
- 2016
- Full Text
- View/download PDF
39. Inflammatory Eicosanoids Increase Amyloid Precursor Protein Expression via Activation of Multiple Neuronal Receptors.
- Author
-
Herbst-Robinson KJ, Liu L, James M, Yao Y, Xie SX, and Brunden KR
- Subjects
- Alzheimer Disease genetics, Alzheimer Disease metabolism, Amyloid beta-Peptides genetics, Amyloid beta-Peptides metabolism, Amyloid beta-Protein Precursor genetics, Animals, Brain pathology, Cells, Cultured, Dinoprostone metabolism, Disease Models, Animal, Female, GTP-Binding Protein alpha Subunits, Gq-G11 genetics, GTP-Binding Protein alpha Subunits, Gq-G11 metabolism, Gene Expression, HEK293 Cells, Humans, Immunoblotting, Inflammation Mediators metabolism, Male, Mice, Transgenic, Protein Kinase C genetics, Protein Kinase C metabolism, RNA Interference, Rats, Receptors, Thromboxane A2, Prostaglandin H2 genetics, Reverse Transcriptase Polymerase Chain Reaction, Thromboxane A2 metabolism, Amyloid beta-Protein Precursor metabolism, Brain metabolism, Eicosanoids metabolism, Neurons metabolism, Receptors, Thromboxane A2, Prostaglandin H2 metabolism
- Abstract
Senile plaques comprised of Aβ peptides are a hallmark of Alzheimer's disease (AD) brain, as are activated glia that release inflammatory molecules, including eicosanoids. Previous studies have demonstrated that amyloid precursor protein (APP) and Aβ levels can be increased through activation of thromboxane A2-prostanoid (TP) receptors on neurons. We demonstrate that TP receptor regulation of APP expression depends on Gαq-signaling and conventional protein kinase C isoforms. Importantly, we discovered that Gαq-linked prostaglandin E2 and leukotriene D4 receptors also regulate APP expression. Prostaglandin E2 and thromboxane A2, as well as total APP levels, were found to be elevated in the brains of aged 5XFAD transgenic mice harboring Aβ plaques and activated glia, suggesting that increased APP expression resulted from eicosanoid binding to Gαq-linked neuronal receptors. Notably, inhibition of eicosanoid synthesis significantly lowered brain APP protein levels in aged 5XFAD mice. These results provide new insights into potential AD therapeutic strategies.
- Published
- 2015
- Full Text
- View/download PDF
40. Pharmacokinetic, pharmacodynamic and metabolic characterization of a brain retentive microtubule (MT)-stabilizing triazolopyrimidine.
- Author
-
Cornec AS, James MJ, Kovalevich J, Trojanowski JQ, Lee VM, Smith AB 3rd, Ballatore C, and Brunden KR
- Subjects
- Animals, Mice, Molecular Conformation, Pyrimidines administration & dosage, Triazoles administration & dosage, Brain metabolism, Microtubules metabolism, Pyrimidines metabolism, Pyrimidines pharmacokinetics, Triazoles metabolism, Triazoles pharmacokinetics
- Abstract
Previous studies revealed that examples of the non-naturally occurring microtubule (MT)-stabilizing triazolopyrimidines are both brain penetrant and orally bioavailable, indicating that this class of compounds may be potentially attractive in the development of MT-stabilizing therapies for the central nervous system (CNS). We now report on the pharmacokinetics (PK), pharmacodynamics (PD), and metabolism of a selected triazolopyrimidine congener, (S)-3-(4-(5-chloro-7-((1,1,1-trifluoropropan-2-yl)amino)-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)-3,5-difluorophenoxy)-propan-1-ol (4). These studies revealed that 4 exhibits longer brain than plasma half-life that may be exploited to achieve a selective accumulation of the compound within the CNS. Furthermore, compound metabolism studies suggest that in plasma 4 is rapidly oxidized at the terminal hydroxyl group to form a comparatively inactive carboxylic acid metabolite. Peripheral administration of relatively low doses of 4 to normal mice was found to produce a significant elevation in acetylated α-tubulin, a marker of stable MTs, in the brain. Collectively, these results indicate that 4 may effectively target brain MTs at doses that produce minimal peripheral exposure., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
41. Passive immunization with phospho-tau antibodies reduces tau pathology and functional deficits in two distinct mouse tauopathy models.
- Author
-
Sankaranarayanan S, Barten DM, Vana L, Devidze N, Yang L, Cadelina G, Hoque N, DeCarr L, Keenan S, Lin A, Cao Y, Snyder B, Zhang B, Nitla M, Hirschfeld G, Barrezueta N, Polson C, Wes P, Rangan VS, Cacace A, Albright CF, Meredith J Jr, Trojanowski JQ, Lee VM, Brunden KR, and Ahlijanian M
- Subjects
- Alzheimer Disease chemically induced, Alzheimer Disease immunology, Alzheimer Disease pathology, Animals, Cerebral Cortex drug effects, Cerebral Cortex immunology, Cerebral Cortex pathology, Cognition Disorders chemically induced, Cognition Disorders immunology, Cognition Disorders pathology, Disease Models, Animal, Exploratory Behavior drug effects, Gene Expression Regulation, Hippocampus drug effects, Hippocampus immunology, Hippocampus pathology, Male, Mice, Mice, Transgenic, Neurons drug effects, Neurons immunology, Neurons pathology, Primary Cell Culture, Recombinant Proteins administration & dosage, Recombinant Proteins adverse effects, Signal Transduction, Treatment Outcome, tau Proteins genetics, tau Proteins immunology, Alzheimer Disease therapy, Antibodies, Monoclonal pharmacology, Cognition Disorders therapy, Immunization, Passive, Phosphoproteins pharmacology, tau Proteins antagonists & inhibitors
- Abstract
In Alzheimer's disease (AD), an extensive accumulation of extracellular amyloid plaques and intraneuronal tau tangles, along with neuronal loss, is evident in distinct brain regions. Staging of tau pathology by postmortem analysis of AD subjects suggests a sequence of initiation and subsequent spread of neurofibrillary tau tangles along defined brain anatomical pathways. Further, the severity of cognitive deficits correlates with the degree and extent of tau pathology. In this study, we demonstrate that phospho-tau (p-tau) antibodies, PHF6 and PHF13, can prevent the induction of tau pathology in primary neuron cultures. The impact of passive immunotherapy on the formation and spread of tau pathology, as well as functional deficits, was subsequently evaluated with these antibodies in two distinct transgenic mouse tauopathy models. The rTg4510 transgenic mouse is characterized by inducible over-expression of P301L mutant tau, and exhibits robust age-dependent brain tau pathology. Systemic treatment with PHF6 and PHF13 from 3 to 6 months of age led to a significant decline in brain and CSF p-tau levels. In a second model, injection of preformed tau fibrils (PFFs) comprised of recombinant tau protein encompassing the microtubule-repeat domains into the cortex and hippocampus of young P301S mutant tau over-expressing mice (PS19) led to robust tau pathology on the ipsilateral side with evidence of spread to distant sites, including the contralateral hippocampus and bilateral entorhinal cortex 4 weeks post-injection. Systemic treatment with PHF13 led to a significant decline in the spread of tau pathology in this model. The reduction in tau species after p-tau antibody treatment was associated with an improvement in novel-object recognition memory test in both models. These studies provide evidence supporting the use of tau immunotherapy as a potential treatment option for AD and other tauopathies.
- Published
- 2015
- Full Text
- View/download PDF
42. Intracerebral injection of preformed synthetic tau fibrils initiates widespread tauopathy and neuronal loss in the brains of tau transgenic mice.
- Author
-
Peeraer E, Bottelbergs A, Van Kolen K, Stancu IC, Vasconcelos B, Mahieu M, Duytschaever H, Ver Donck L, Torremans A, Sluydts E, Van Acker N, Kemp JA, Mercken M, Brunden KR, Trojanowski JQ, Dewachter I, Lee VM, and Moechars D
- Subjects
- Age Factors, Analysis of Variance, Animals, Disease Models, Animal, Disease Progression, Functional Laterality, Gene Expression Regulation drug effects, Gene Expression Regulation genetics, Hippocampus drug effects, Hippocampus metabolism, Hippocampus pathology, Humans, Mice, Mice, Transgenic, Mutation genetics, Neurofibrillary Tangles metabolism, tau Proteins chemistry, Tauopathies chemically induced, Tauopathies genetics, Tauopathies pathology, tau Proteins administration & dosage, tau Proteins genetics
- Abstract
Neurofibrillary tangles composed of hyperphosphorylated fibrillized tau are found in numerous tauopathies including Alzheimer's disease. Increasing evidence suggests that tau pathology can be transmitted from cell-to-cell; however the mechanisms involved in the initiation of tau fibrillization and spreading of disease linked to progression of tau pathology are poorly understood. We show here that intracerebral injections of preformed synthetic tau fibrils into the hippocampus or frontal cortex of young tau transgenic mice expressing mutant human P301L tau induces tau hyperphosphorylation and aggregation around the site of injection, as well as a time-dependent propagation of tau pathology to interconnected brain areas distant from the injection site. Furthermore, we show that the tau pathology as a consequence of injection of tau preformed fibrils into the hippocampus induces selective loss of CA1 neurons. Together, our data confirm previous studies on the seeded induction and the spreading of tau pathology in a different tau transgenic mouse model and reveals neuronal loss associated with seeded tau pathology in tau transgenic mouse brain. These results further validate the utility of the tau seeding model in studying disease transmission, and provide a more complete in vivo tauopathy model with associated neurodegeneration which can be used to investigate the mechanisms involved in tau aggregation and spreading, as well as aid in the search for disease modifying treatments for Alzheimer's disease and related tauopathies., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
43. Microtubule-stabilizing agents as potential therapeutics for neurodegenerative disease.
- Author
-
Brunden KR, Trojanowski JQ, Smith AB 3rd, Lee VM, and Ballatore C
- Subjects
- Animals, Humans, Microtubules metabolism, Molecular Structure, Neurodegenerative Diseases metabolism, Structure-Activity Relationship, Tubulin Modulators chemistry, Microtubules drug effects, Neurodegenerative Diseases drug therapy, Tubulin Modulators pharmacology, Tubulin Modulators therapeutic use
- Abstract
Microtubules (MTs), cytoskeletal elements found in all mammalian cells, play a significant role in cell structure and in cell division. They are especially critical in the proper functioning of post-mitotic central nervous system neurons, where MTs serve as the structures on which key cellular constituents are trafficked in axonal projections. MTs are stabilized in axons by the MT-associated protein tau, and in several neurodegenerative diseases, including Alzheimer's disease, frontotemporal lobar degeneration, and Parkinson's disease, tau function appears to be compromised due to the protein dissociating from MTs and depositing into insoluble inclusions referred to as neurofibrillary tangles. This loss of tau function is believed to result in alterations of MT structure and function, resulting in aberrant axonal transport that likely contributes to the neurodegenerative process. There is also evidence of axonal transport deficiencies in other neurodegenerative diseases, including amyotrophic lateral sclerosis and Huntington's disease, which may result, at least in part, from MT alterations. Accordingly, a possible therapeutic strategy for such neurodegenerative conditions is to treat with MT-stabilizing agents, such as those that have been used in the treatment of cancer. Here, we review evidence of axonal transport and MT deficiencies in a number of neurodegenerative diseases, and summarize the various classes of known MT-stabilizing agents. Finally, we highlight the growing evidence that small molecule MT-stabilizing agents provide benefit in animal models of neurodegenerative disease and discuss the desired features of such molecules for the treatment of these central nervous system disorders., (Copyright © 2014 Elsevier Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
44. Evaluation of the cyclopentane-1,2-dione as a potential bio-isostere of the carboxylic acid functional group.
- Author
-
Ballatore C, Gay B, Huang L, Robinson KH, James MJ, Trojanowski JQ, Lee VM, Brunden KR, and Smith AB 3rd
- Subjects
- Carboxylic Acids chemical synthesis, Carboxylic Acids chemistry, Crystallography, X-Ray, Cyclopentanes chemical synthesis, Cyclopentanes chemistry, Dose-Response Relationship, Drug, Humans, Models, Molecular, Molecular Structure, Structure-Activity Relationship, Sulfonamides chemical synthesis, Sulfonamides chemistry, Carboxylic Acids pharmacology, Cyclopentanes pharmacology, Receptors, Thromboxane A2, Prostaglandin H2 antagonists & inhibitors, Sulfonamides pharmacology
- Abstract
Cycloalkylpolyones hold promise in drug design as carboxylic acid bio-isosteres. To investigate cyclopentane-1,2-diones as potential surrogates of the carboxylic acid functional group, the acidity, tautomerism, and geometry of hydrogen bonding of representative compounds were evaluated. Prototypic derivatives of the known thromboxane A2 prostanoid (TP) receptor antagonist, 3-(3-(2-((4-chlorophenyl)sulfonamido)-ethyl)phenyl)propanoic acid, in which the carboxylic acid moiety is replaced by the cyclopentane-1,2-dione unit, were synthesized and evaluated as TP receptor antagonists. Cyclopentane-1,2-dione derivative 9 was found to be a potent TP receptor antagonist with an IC50 value comparable to that of the parent carboxylic acid. These results indicate that the cyclopentane-1,2-dione may be a potentially useful carboxylic acid bio-isostere., (Copyright © 2014 Elsevier Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
45. Brain-penetrant, orally bioavailable microtubule-stabilizing small molecules are potential candidate therapeutics for Alzheimer's disease and related tauopathies.
- Author
-
Lou K, Yao Y, Hoye AT, James MJ, Cornec AS, Hyde E, Gay B, Lee VM, Trojanowski JQ, Smith AB 3rd, Brunden KR, and Ballatore C
- Subjects
- Administration, Oral, Alzheimer Disease drug therapy, Alzheimer Disease pathology, Animals, Biological Availability, Cell Line, Humans, Mice, Microtubules metabolism, Molecular Structure, Pyrimidines administration & dosage, Pyrimidines chemistry, Tauopathies pathology, Brain metabolism, Microtubules drug effects, Pyrimidines pharmacology, Tauopathies drug therapy
- Abstract
Microtubule (MT) stabilizing drugs hold promise as potential treatments for Alzheimer's disease (AD) and related tauopathies. However, thus far epothilone D has been the only brain-penetrant MT-stabilizer to be evaluated in tau transgenic mice and in AD patients. Furthermore, this natural product exhibits potential deficiencies as a drug candidate, including an intravenous route of administration and the inhibition of the P-glycoprotein (Pgp) transporter. Thus, the identification of alternative CNS-active MT-stabilizing agents that lack these potential limitations is of interest. Toward this objective, we have evaluated representative compounds from known classes of non-naturally occurring MT-stabilizing small molecules. This led to the identification of selected triazolopyrimidines and phenylpyrimidines that are orally bioavailable and brain-penetrant without disruption of Pgp function. Pharmacodynamic studies confirmed that representative compounds from these series enhance MT-stabilization in the brains of wild-type mice. Thus, these classes of MT-stabilizers hold promise for the development of orally active, CNS-directed MT-stabilizing therapies.
- Published
- 2014
- Full Text
- View/download PDF
46. Potent, long-acting cyclopentane-1,3-Dione thromboxane (A2)-receptor antagonists.
- Author
-
Wang X, Liu L, Huang L, Herbst-Robinson K, Cornec AS, James MJ, Sugiyama S, Bassetto M, Brancale A, Trojanowski JQ, Lee VM, Smith AB 3rd, Brunden KR, and Ballatore C
- Abstract
A series of derivatives of the known thromboxane A2 prostanoid (TP) receptor antagonists, 3-(6-((4-chlorophenyl)sulfonamido)-5,6,7,8-tetrahydronaphthalen-1-yl)propanoic acid and 3-(3-(2-((4-chlorophenyl)sulfonamido)ethyl)phenyl) propanoic acid, were synthesized in which the carboxylic acid functional group was replaced with substituted cyclopentane-1,3-dione (CPD) bioisosteres. Characterization of these molecules led to the discovery of remarkably potent new analogues, some of which were considerably more active than the corresponding parent carboxylic acid compounds. Depending on the choice of the C2 substituent of the CPD unit, these new derivatives can produce either a reversible or an apparent irreversible inhibition of the human TP receptor. Given the potency and the long-lasting inhibition of TP receptor signaling, these novel antagonists may comprise promising leads for the development of antithromboxane therapies.
- Published
- 2014
- Full Text
- View/download PDF
47. Aminothienopyridazines as imaging probes of tau pathology: a patent evaluation of WO2013090497.
- Author
-
Ballatore C, Smith AB 3rd, Lee VM, Trojanowski JQ, and Brunden KR
- Subjects
- Alzheimer Disease metabolism, Alzheimer Disease pathology, Animals, Biomarkers metabolism, Brain metabolism, Brain pathology, Humans, Patents as Topic, Predictive Value of Tests, Alzheimer Disease diagnostic imaging, Brain diagnostic imaging, Molecular Imaging methods, Positron-Emission Tomography, Pyridazines metabolism, Pyridazines pharmacokinetics, Radiopharmaceuticals metabolism, Radiopharmaceuticals pharmacokinetics, tau Proteins metabolism
- Abstract
Small-molecule ligands, amenable to positron emission tomography (PET) imaging of different types of neurodegenerative disease-associated amyloid deposits in the CNS of living patients, hold considerable promise for diagnostic purposes, as well as for monitoring disease progression and the effectiveness of treatments. The patent entitled 'Heterocyclic Compounds as Imaging Probes of Tau Pathology' (WO2013090497) discloses the identification of a novel class of tau imaging agents, the aminothienopyridazines. Selected compounds from this class are described that can stain selectively tau pathology in brain tissue sections. Moreover, examples of this class of compounds exhibit absorption, distribution, metabolism, excretion and pharmacokinetics (ADME-PK) properties that are appropriate for CNS PET ligands.
- Published
- 2014
- Full Text
- View/download PDF
48. Design, synthesis, and biological evaluation of 1-phenylpyrazolo[3,4-e]pyrrolo[3,4-g]indolizine-4,6(1H,5H)-diones as new glycogen synthase kinase-3β inhibitors.
- Author
-
La Pietra V, La Regina G, Coluccia A, Famiglini V, Pelliccia S, Plotkin B, Eldar-Finkelman H, Brancale A, Ballatore C, Crowe A, Brunden KR, Marinelli L, Novellino E, and Silvestri R
- Subjects
- Cell Line, Dose-Response Relationship, Drug, Glycogen Synthase Kinase 3 metabolism, Glycogen Synthase Kinase 3 beta, Humans, Indolizidines chemical synthesis, Indolizidines chemistry, Models, Molecular, Molecular Structure, Protein Kinase Inhibitors chemistry, Recombinant Proteins metabolism, Structure-Activity Relationship, Succinimides chemical synthesis, Succinimides chemistry, Drug Design, Glycogen Synthase Kinase 3 antagonists & inhibitors, Indolizidines pharmacology, Protein Kinase Inhibitors chemical synthesis, Protein Kinase Inhibitors pharmacology, Succinimides pharmacology
- Abstract
Compound 5 was selected from our in-house library as a suitable starting point for the rational design of new GSK-3β inhibitors. MC/FEP calculations of 5 led to the identification of a structural class of new GSK-3β inhibitors. Compound 18 inhibited GSK-3β with an IC50 of 0.24 μM and inhibited tau phosphorylation in a cell-based assay. It proved to be a selective inhibitor of GSK-3 against a panel of 17 kinases and showed >10-fold selectivity against CDK2. Calculated physicochemical properties and Volsurf predictions suggested that compound 18 has the potential to diffuse passively across the blood-brain barrier.
- Published
- 2013
- Full Text
- View/download PDF
49. MT-Stabilizer, Dictyostatin, Exhibits Prolonged Brain Retention and Activity: Potential Therapeutic Implications.
- Author
-
Brunden KR, Gardner NM, James MJ, Yao Y, Trojanowski JQ, Lee VM, Paterson I, Ballatore C, and Smith AB 3rd
- Abstract
Inclusions comprising the microtubule (MT)-stabilizing protein, tau, are found within neurons in the brains of patients with Alzheimer's disease and related neurodegenerative disorders that are broadly referred to as tauopathies. The sequestration of tau into inclusions is believed to cause a loss of tau function, such that MT structure and function are compromised, leading to neuronal damage. Recent data reveal that the brain-penetrant MT-stabilizing agent, epothilone D (EpoD), improves cognitive function and decreases both neuron loss and tau pathology in transgenic mouse models of tauopathy. There is thus a need to identify additional MT-stabilizing compounds with blood-brain barrier (BBB) permeability and slow brain clearance, as observed with EpoD. We report here that the MT-stabilizing natural product, dictyostatin, crosses the BBB in mice and has extended brain retention. Moreover, a single administration of dictyostatin to mice causes prolonged stabilization of MTs in the brain. In contrast, the structurally related MT-stabilizer, discodermolide, shows significantly less brain exposure. Thus, dictyostatin merits further investigation as a potential tauopathy therapeutic.
- Published
- 2013
- Full Text
- View/download PDF
50. Lewy body-like α-synuclein aggregates resist degradation and impair macroautophagy.
- Author
-
Tanik SA, Schultheiss CE, Volpicelli-Daley LA, Brunden KR, and Lee VM
- Subjects
- Animals, HEK293 Cells, HeLa Cells, Humans, Lewy Bodies genetics, Lewy Bodies pathology, Mice, Neurons pathology, Parkinson Disease genetics, Parkinson Disease pathology, Proteasome Endopeptidase Complex genetics, Proteasome Endopeptidase Complex metabolism, alpha-Synuclein genetics, Autophagy, Lewy Bodies metabolism, Models, Biological, Neurons metabolism, Parkinson Disease metabolism, Proteolysis, alpha-Synuclein metabolism
- Abstract
Cytoplasmic α-synuclein (α-syn) aggregates, referred to as Lewy bodies, are pathological hallmarks of a number of neurodegenerative diseases, most notably Parkinson disease. Activation of macroautophagy is suggested to facilitate degradation of certain proteinaceous inclusions, but it is unclear if this pathway is capable of degrading α-syn aggregates. Here, we examined this issue by utilizing cellular models in which intracellular Lewy body-like α-syn inclusions accumulate after internalization of pre-formed α-syn fibrils into α-syn-expressing HEK293 cells or cultured primary neurons. We demonstrate that α-syn inclusions cannot be effectively degraded, even though they co-localize with essential components of both the autophagic and proteasomal protein degradation pathways. The α-syn aggregates persist even after soluble α-syn levels have been substantially reduced, suggesting that once formed, the α-syn inclusions are refractory to clearance. Importantly, we also find that α-syn aggregates impair overall macroautophagy by reducing autophagosome clearance, which may contribute to the increased cell death that is observed in aggregate-bearing cells.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.