Cathepsin B in neurodegeneration of Alzheimer's disease, traumatic brain injury, and related brain disorders

https://doi.org/10.1016/j.bbapap.2020.140428Get rights and content

Highlights

  • Cathepsin B is elevated in human Alzheimer's disease (AD) and traumatic brain injury (TBI) patients, and correlates with behavioral and injury outcomes

  • Inhibition of cathepsin B in AD and TBI animal models results in alleviation of cognitive and behavioral deficits with improvement in neuropathology

  • Preprocathepsin B undergoes processing to generate mature cathepsin B, which can be selectively inhibited by CA-074

  • The occluding loop of cathepsin B regulates dipeptidylcarboxypeptidase and endopeptidase activities of cathepsin B

  • Lysosomal leakage of cathepsin B to the cytosol participates in cell death and inflammation of AD, TBI, and related brain disorders

Abstract

Investigations of Alzheimer's disease (AD), traumatic brain injury (TBI), and related brain disorders have provided extensive evidence for involvement of cathepsin B, a lysosomal cysteine protease, in mediating the behavioral deficits and neuropathology of these neurodegenerative diseases. This review integrates findings of cathepsin B regulation in clinical biomarker studies, animal model genetic and inhibitor evaluations, structural studies, and lysosomal cell biological mechanisms in AD, TBI, and related brain disorders. The results together indicate the role of cathepsin B in the behavioral deficits and neuropathology of these disorders. Lysosomal leakage occurs in AD and TBI, and related neurodegeneration, which leads to the hypothesis that cathepsin B is redistributed from the lysosome to the cytosol where it initiates cell death and inflammation processes associated with neurodegeneration. These results together implicate cathepsin B as a major contributor to these neuropathological changes and behavioral deficits. These findings support the investigation of cathepsin B as a potential drug target for therapeutic discovery and treatment of AD, TBI, and TBI-related brain disorders.

Introduction

Neurodegeneration of Alzheimer's disease (AD), traumatic brain injury (TBI), and related dementia (ADRD) disorders displays behavioral deficits in memory, cognition, problem solving, executive function, language, emotion, and related brain functions [[1], [2], [3], [4], [5], [6], [7], [8], [9], [10], [11], [12], [13], [14], [15]]. Severe cognitive decline, dementia, occurs in ADRD disorders of Alzheimer's disease [[1], [2], [3]], traumatic brain injury (TBI) [[4], [5], [6]], brain ischemia and stroke [[7], [8], [9]], and related neurodegenerative brain disorders [[10], [11], [12], [13], [14], [15]]. These disorders are widespread affecting more than 47 million people worldwide, with high prevalence in the aged population.

There have been tremendous efforts in the field of drug discovery for AD, TBI, and numerous related ADRD disorders, but decades of drug development research have not yet yielded effective therapeutics to attenuate or reverse these neurodegenerative diseases. Advancements in dementia therapeutics strategies will necessitate elucidation of novel molecular mechanisms in ADRD which can be modulated through drug intervention to ameliorate the behavioral dysfunctions and neuropathology of ADRD disorders. Investigation of novel targets of ADRD will advance opportunities for therapeutics development to extend prior efforts for targeting neuropathological amyloid-β (Aβ) [[16], [17], [18], [19], [20]], phospho-tau [[21], [22], [23]], and related mechanisms [[24], [25], [26], [27]].

Cathepsin B, a lysosomal cysteine protease, represents a candidate drug target for AD, TBI, and related brain disorders. Cathepsin B has been shown by numerous studies to participate in AD and TBI cognitive and behavioral deficits, and in neuropathology. This review highlights the multi-disciplinary research of cathepsin B in human clinical studies, and in AD, TBI, and related animal models using cathepsin B gene knockout and chemical inhibition. Findings from these studies support the hypothesis that lysosomal leakage of cathepsin B to the cytosol leads to neurodegeneration and behavioral deficits in AD, TBI, and related brain disorders. These results provide the rationale for investigation of cathepsin B as a mechanistic drug target for therapeutics discovery and development for AD, TBI, and related dementia.

Section snippets

Cathepsin B elevation in human clinical AD, TBI, and related neurodegenerative disorders

Several studies have shown increased levels of cathepsin B protein or activity in plasma, CSF, and amyloid plaques in human AD, TBI, and related neurodegenerative diseases (Table 1) [[28], [29], [30], [31], [32], [33], [34], [35], [36], [37], [38], [39]]. Importantly, increased plasma cathepsin B correlates with cognitive dysfunction in AD [29] and severe trauma outcomes [36].

Human brain gene expression of cathepsin B at young to adult ages

Analyses of cathepsin B expression in human brain illustrates its abundant expression in hippocampal and cortical regions which are responsible for cognitive and behavioral functions [40]. Cathepsin B mRNA expression is illustrated as RNAseq data overlaid on a human brain MRI image (from the Allen human brain atlas, https://human.brain-map.org/) [41,42] in Fig. 2a.

Cathepsin B is one of the most abundant lysosomal cathepsin proteases expressed in human brain compared to the 15 member lysosomal

Cathepsin B participates in behavioral deficits and neuropathology in animal models of AD, TBI, and related brain disorders

Numerous investigations of animal models of AD, TBI, and related dementia disorders demonstrate that cathepsin B is elevated during brain dysfunction and pathogenesis [29,[44], [45], [46], [47], [48], [49], [50], [51], [52], [53], [54], [55], [56], [57], [58], [59], [60], [61], [62], [63], [64], [65], [66], [67], [68], [69], [70], [71], [72], [73], [74]]. Furthermore, gene knockout of cathepsin B in these animal models of brain disorders results in amelioration of behavioral deficits and

Maturation of preprocathepsin B, member of the cysteine cathepsin protease family

Cathepsin B is a lysosomal cysteine protease belonging to the clan CA and the papain-like family C1A. The other C1A papain-like cysteine proteases in humans are cathepsins C, F, H, K, L, O, S, V, W, and Z (also known as cathepsin X) [40,43,114]. The cathepsin family also includes the aspartyl proteases cathepsins D and E, and the serine proteases cathepsins A and G. These cathepsins comprise the 15 protease members of lysosomal cathepsin proteases. Cathepsin B is among the most abundantly

Selective CA-074 inhibitor of cathepsin B

Development of inhibitors of cathepsin B may lead to novel therapeutic agents for the treatment of the devastating AD, TBI, and related cognitive and behavioral deficits. Selectivity of inhibitors for cathepsin B compared to other members of the cysteine cathepsin family is important for targeted inhibition of cathepsin B without disturbing other related proteases and associated functions. Distinct structural features of cathepsin B have resulted in design and synthesis of the highly selective

Regulation of cathepsin B proteolysis by its distinct occluding loop

Cathepsin B uniquely possesses the occluding loop compared to other cysteine cathepsins [[132], [133], [134], [135], [136], [137]]. The occluding loop is located near the enzyme's substrate binding pocket containing the active site Cys-29 residue [100,132] (Fig. 5) which catalyzes endopeptidase and dipeptidyl carboxypeptidase activities. Thus, these activities are influenced by the occluding situated near the primed subsites which favor binding of peptide substrates with two residues

Lysosomal leakage of cathepsin B to the cytosol in cell death and inflammation of AD, TBI, and related brain disorders

Abnormal lysosomal leakage of cathepsin B into the cytosol of cells occurs as an integral mechanism for cell death and inflammation in AD, TBI, and related brain disorders. Cathepsin B and cathepsin proteases are normally sequestered within lysosome organelles for homeostasis by degradation of cellular proteins into amino acids [138,139]. In AD, TBI, and related brain disorders, lysosomal membranes lose their integrity to result in lysosomal leakage and redistribution of cathepsin B to the

Translocation of lysosomal cathepsin B to the cytosol in neurodegeneration

Lysosomal leakage results in the pathogenic redistribution of cathepsin B from the lysosome to the cytosol in neurodegeneration, in contrast to the normal intra-lysosomal location of cathepsin B for protein degradation and homeostasis, which leads to the hypothesis that abnormal cytosolic cathepsin B participates in AD and TBI-related dysfunctions (illustrated in Fig. 6). The key question is what are the cytosolic substrates of cathepsin B that lead to neurodegeneration? The profiles of

Conclusions and future perspectives

The hypothesis that lysosomal leakage of pathogenic cathepsin B to the cytosol results in neurodegeneration and behavioral deficits of AD, TBI, and related brain disorders (Fig. 6) is supported by ample evidence from clinical and animal model studies of AD, TBI, and related brain disorders. This leads to the question of what are the cytosolic substrates of cathepsin B that lead to neurodegeneration? Current data implicate cathepsin B substrates involved in cell death and inflammatory pathways.

Declaration of Competing Interest

None

Acknowledgments

This work was supported by NIH grant R01NS109075 to VH, and NIH grant R41NS110147 to GH. The authors thank Michael Gilson and the Center for Drug Discovery Innovation (cDDI) at UC San Diego for advice and resources for the Molecular Operating Environment (MOE) analyses.

V. Hook organized the manuscript for participation by coauthors and wrote the manuscript. MY, CM, and AJO conducted the MOE representation of the cathepsin B structure complexed with the CA-074 inhibitors, and conducted

References (197)

  • K. Tsuchiya et al.

    Postictal blockade of ischemic hippocampal neuronal death in primates using selective cathepsin inhibitors

    Exp. Neurol.

    (1999 Feb)
  • Z. Wu et al.

    Cathepsin B plays a critical role in inducing Alzheimer's disease-like phenotypes following chronic systemic exposure to lipopolysaccharide from Porphyromonas gingivalis in mice

    Brain Behav. Immun.

    (2017 Oct)
  • V.Y. Hook et al.

    Genetic cathepsin B deficiency reduces beta-amyloid in transgenic mice expressing human wild-type amyloid precursor protein

    Biochem. Biophys. Res. Commun.

    (2009 Aug 21)
  • T.C. Saido et al.

    Dominant and differential deposition of distinct beta-amyloid peptide species, A beta N3(pE), in senile plaques

    Neuron.

    (1995 Feb)
  • M.L. Wang et al.

    Cognitive impairment after traumatic brain injury: The role of MRI and possible pathological basis

    J. Neurol. Sci.

    (2016 Nov 15)
  • J. Julien et al.

    Association of traumatic brain injury and Alzheimer disease onset: a systematic review

    Ann. Phys. Rehabil. Med.

    (2017 Sep)
  • J.W. Lee et al.

    Neuro-inflammation induced by lipopolysaccharide causes cognitive impairment through enhancement of beta-amyloid generation

    J. Neuroinflammation

    (2008 Aug 29)
  • V.Y. Hook et al.

    Inhibitors of cathepsin B improve memory and reduce beta-amyloid in transgenic Alzheimer disease mice expressing the wild-type, but not the Swedish mutant, beta-secretase site of the amyloid precursor protein

    J. Biol. Chem.

    (2008 Mar 21)
  • M. Murata et al.

    Novel epoxysuccinyl peptides. Selective inhibitors of cathepsin B, in vitro

    FEBS Lett.

    (1991 Mar 25)
  • T. Towatari et al.

    Novel epoxysuccinyl peptides. A selective inhibitor of cathepsin B, in vivo

    FEBS Lett.

    (1991 Mar 25)
  • T.J. Montine et al.

    Recommendations of the Alzheimer's disease-related dementias conference

    Neurology.

    (2014 Aug 26)
  • R.A. Corriveau et al.

    Alzheimer's disease-related dementias summit 2016: national research priorities

    Neurology.

    (2017 Dec 5)
  • P.S. Aisen et al.

    On the path to 2025: understanding the Alzheimer's disease continuum

    Alzheimers Res. Ther.

    (2017 Aug 9)
  • J. Ramalho et al.

    Dementia resulting from traumatic brain injury

    Dement Neuropsychol.

    (2015 Oct-Dec)
  • C. LoBue et al.

    Traumatic brain injury and risk of long-term brain changes, accumulation of pathological markers, and developing dementia: a review

    J. Alzheimers Dis.

    (2019)
  • M.D. Mijajlović et al.

    Post-stroke dementia - a comprehensive review

    BMC Med.

    (2017 Jan 18)
  • P. Corraini et al.

    Long-term risk of dementia among survivors of ischemic or hemorrhagic stroke

    Stroke.

    (2017 Jan)
  • A. Charidimou et al.

    International META-MICROBLEEDS Initiative. Clinical significance of cerebral microbleeds on MRI: A comprehensive meta-analysis of risk of intracerebral hemorrhage, ischemic stroke, mortality, and dementia in cohort studies (v1)

    Int. J. Stroke

    (2018 Jul)
  • J. Gratwicke et al.

    Parkinson's disease dementia: a neural networks perspective

    Brain.

    (2015 Jun)
  • S.N. Gomperts

    Lewy body dementias: dementia with lewy bodies and Parkinson disease dementia

    Continuum (Minneap Minn)

    (2016 Apr)
  • E.L. Whitlock et al.

    Association between persistent pain and memory decline and dementia in a longitudinal cohort of elders

    JAMA Intern. Med.

    (2017 Aug 1)
  • J. Hardy et al.

    The amyloid hypothesis of Alzheimer's disease: progress and problems on the road to therapeutics

    Science.

    (2002 Jul 19)
  • R.A. Sperling et al.

    Testing the right target and right drug at the right stage

    Sci. Transl. Med.

    (2011 Nov)
  • S. Schilling et al.

    Passive Aβ immunotherapy: current achievements and future perspectives

    Molecules

    (2018 May 3)
  • R. Ricciarelli et al.

    The amyloid cascade hypothesis in Alzheimer's disease: it's time to change our mind

    Curr. Neuropharmacol.

    (2017)
  • C. Li et al.

    Tau-based therapies in neurodegeneration: opportunities and challenges

    Nat. Rev. Drug Discov.

    (2017 Dec)
  • E.E. Congdon et al.

    Tau-targeting therapies for Alzheimer disease

    Nat. Rev. Neurol.

    (2018 Jul)
  • J. Cummings et al.

    Drug development in Alzheimer's disease: the path to 2025

    Alzheimers Res. Ther.

    (2016 Sep 20)
  • R. Loera-Valencia et al.

    Current and emerging avenues for Alzheimer's disease drug targets

    J. Intern. Med.

    (2019 Oct)
  • P.M. Kochanek et al.

    Paths to successful translation of new therapies for severe traumatic brain injury in the golden age of traumatic brain injury research: a Pittsburgh vision

    J. Neurotrauma

    (2019)
  • J. Sundelöf et al.

    Higher cathepsin B levels in plasma in Alzheimer's disease compared to healthy controls

    J. Alzheimers Dis.

    (2010)
  • Y. Sun et al.

    Translational study of Alzheimer's disease (AD) biomarkers from brain tissues in AβPP/PS1 mice and serum of AD patients

    J. Alzheimers Dis.

    (2015)
  • F. Morena et al.

    A comparison of lysosomal enzymes expression levels in peripheral blood of mild- and severe-Alzheimer's disease and MCI patients: implications for regenerative medicine approaches

    Int. J. Mol. Sci.

    (2017 Aug 19)
  • A.M. Cataldo et al.

    Enzymatically active lysosomal proteases are associated with amyloid deposits in Alzheimer brain

    Proc. Natl. Acad. Sci. U. S. A.

    (1990 May)
  • J. Zhang et al.

    Quantitative proteomics of cerebrospinal fluid from patients with Alzheimer disease. J Alzheimers Dis. 2005 Apr;7(2):125-33; discussion 173-80

    Erratum in: J. Alzheimers Dis.

    (2006 Mar)
  • Y. Hou et al.

    Ageing as a risk factor for neurodegenerative disease

    Nat. Rev. Neurol.

    (2019 Oct)
  • Y. Hara et al.

    Translating the biology of aging into novel therapeutics for Alzheimer disease

    Neurology.

    (2019 Jan 8)
  • E. Nilsson et al.

    Cerebrospinal fluid cathepsin B and S

    Neurol. Sci.

    (2013 Apr)
  • I. Assfalg-Machleidt et al.

    Cathepsin B-indicator for the release of lysosomal cysteine proteinases in severe trauma and inflammation

    Biol. Chem. Hoppe Seyler

    (1990 May)
  • E. Neugebauer et al.

    Handbook of Mediators in Septic Shock

    (1993)
  • Cited by (85)

    View all citing articles on Scopus
    View full text