Next Article in Journal
Crystal Structures of Bacterial Pectin Methylesterases Pme8A and PmeC2 from Rumen Butyrivibrio
Next Article in Special Issue
Advancements in the Application of Nanomedicine in Alzheimer’s Disease: A Therapeutic Perspective
Previous Article in Journal
Single-Cell RNA-Sequencing Reveals Peripheral T Helper Cells Promoting the Development of IgG4-Related Disease by Enhancing B Cell Activation and Differentiation
Previous Article in Special Issue
α-Synuclein Strains and Their Relevance to Parkinson’s Disease, Multiple System Atrophy, and Dementia with Lewy Bodies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Neurodegenerative Diseases: Molecular Mechanisms and Therapies

by
Zhi Dong Zhou
1,2,* and
Alexandre Hiroaki Kihara
3,4,*
1
National Neuroscience Institute of Singapore, 11 Jalan Tan Tock Seng, Singapore 30843, Singapore
2
Signature Research Program in Neuroscience and Behavioral Disorders, Duke-NUS Graduate Medical School Singapore, 8 College Road, Singapore 169857, Singapore
3
Neurogenetics Laboratory, Universidade Federal do ABC, São Bernardo do Campo 09606-045, SP, Brazil
4
Center for Mathematics, Computing and Cognition, Universidade Federal do ABC, São Bernardo do Campo 09606-045, SP, Brazil
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(18), 13721; https://doi.org/10.3390/ijms241813721
Submission received: 23 August 2023 / Accepted: 4 September 2023 / Published: 6 September 2023
(This article belongs to the Special Issue Neurodegenerative Diseases: Molecular Mechanisms and Therapies)
Neurodegenerative diseases are characterized by the progressive degeneration or death of neurons in the central or peripheral nervous system. Genetic, environmental, and lifestyle factors all contribute to neurodegenerative diseases [1,2]. Although some neurodegenerative diseases can be managed with treatments, and the progression of the disease may be slow, many remain incurable [3]. Common underlying processes contribute to the degeneration of neurons, but the molecular mechanisms of neurodegenerative diseases are complex and varied, and they can differ depending on the specific conditions [4]. Neurodegenerative processes and neuronal disorders also occur in the spinal cord [5], retina [6], and enteric nervous system [7], eventually reflecting and/or affecting what occurs in the brain. The consequences of these conditions are often a gradual decline and abnormality in perceptual, cognitive, motor, behavioral, and social abilities. Neurodegenerative diseases pose a significant public health threat. These diseases commonly occur in elderly individuals and their prevalence increases with age. The increasing lifespan and decreasing fertility rate lead to an increase in the median age prevalence of these disorders and aggravated health and economic burdens to society.
Long-lived people have a higher concentration of the longevity-associated variant (LAV) homozygous genotype in member 4 of the bactericidal/permeability-increasing fold-containing family B (BPIFB4) gene [8]. It was previously shown that LAV-BPIFB4 prevented Huntington’s disease (HD) progression in mice [9]. The production of mutant Huntingtin protein, which exhibits pathological lengthy polyglutamine repeats and toxic consequences, results in HD. In this context, there is a substantial clinical need yet unmet, as there are currently no clinically validated treatment medicines or therapies that can slow or stop neurodegeneration and disease progression in HD. In this Special Issue, Cattaneo et al. demonstrate that LAV-BPIFB4 generates neuroprotection in the striatum-derived STHdh cell line, an in vitro model of HD [10].
The use of mutant cell lines enables the study of the underlying pathophysiological processes causing neurodegeneration, such as oxidative stress. Besides the NADPH oxidase 2 (NOX2/gp91) gene [11,12], other NADPH oxidase family members (NOX 1, 3–5, DUOX1, and DUOX2) have been implicated in the production of reactive oxygen species (ROS) [13]. Interestingly, the NADPH oxidase organizer 1 (NOXO1) is essential for the organization and formation of the NOX1-dependent NADPH oxidase complex. Benssouina et al. demonstrate that, in a mutated NOXO1 colorectal cancer cell line with mutated D-box, there is an increase in ROS production and cytotoxicity through NOX1 activity, affecting the mitochondrial organization. These effects were observed with the translocation of the mutated protein from the soluble membrane fraction to a cytoskeletal insoluble fraction [14]. The D-box of NOXO1 protein seems to maintain the balanced distribution of Noxo1 between membrane and cytoskeleton, which is vital to Nox1-dependent NADPH oxidase activity, ROS generation, and cytotoxicity.
Another line of research that involves the use of in vitro cell lines is related to Parkinson’s disease (PD). In a familial type of PD, loss-of-function mutations of the E3 ligase Parkin likely result in the accumulation of aberrant mitochondria and neurodegeneration, due to impaired polyubiquitination of defective mitochondria for subsequent mitophagy clearance. However, researchers’ attention has recently been directed towards Parkin’s role as a redox molecule. Using cell culture, Ardah et al. overexpressed several combinations of Parkin and its substrates FAF1, PINK1, and ubiquitin to ascertain the function of Parkin as a redox molecule in the mitochondria [15]. Unexpectedly, these authors found that the Parkin monomer was not recruited to aberrant mitochondria but instead self-aggregated into the inner and outer membranes of the mitochondria, thus becoming insoluble. Parkin overexpression alone produced Parkin protein aggregates without self-ubiquitination, which triggered autophagy to clear away damaged mitochondria. They concluded that the polyubiquitination of Parkin substrates on the mitochondria is not indispensable for the mitophagy clearance of defective mitochondria.
Indeed, PD pathophysiology has garnered attention in the scientific community, with recent reports generating original research and reviews. It is established that α-synuclein (αSyn) aggregates and Lewy body formation are some of the pathological features of PD. Regarding pathological features beyond those associated with PD, Graves et al. examined the reciprocal interaction between conformational strains of Syn aggregates and the cellular environment, discussing recent evidence for its participation in other neurodegenerative diseases [16]. The pathological aggregation of αSyn protein has also been identified in dementia with Lewy bodies (DLB), multiple system atrophy (MSA), Alzheimer’s disease (AD), brain traumatic injuries, and neurodegeneration with brain iron accumulation (NBIA). Diseases associated with αSyn protein aggregation are referred to as synucleinopathies, highlighting the potential common pathological events and disease mechanisms in these disorders.
A crucial element in the pathophysiology of PD is the self-association of amylogenic proteins to form fibril structures. Recent studies have suggested the interruption of fibrils with an external electric field as a non-invasive strategy with a few advantages over other techniques for potential clinical applications. Razzokov et al. performed molecular dynamics (MD) simulations on extremely hazardous α-synuclein fibrils to obtain a molecular-level understanding of fibril disruption mechanisms. Their findings revealed that the applied external electric field significantly alters the structure of αsyn fibrils, in addition to highlighting the critical electric field intensity at which the hydrophobic core of αsyn fibrils can be fully opened [17].
The human cystatin C (HCC) amyloidogenic protein uses a domain-swapping process to produce dimers and higher oligomers such as trimers, tetramers, and others. Wojciechowska et al. characterized the HCC oligomeric form observed in solutions with pH levels ranging from 2.2 to 10.0 and in environments that encouraged oligomerization. Size exclusion chromatography, dynamic light scattering, and small-angle X-ray scattering were used to characterize the oligomeric forms of HCC generated under various conditions. It was found that HCC has a strong capacity to form dimers at pH 4.0–5.0 and tetramers at lower pH (2.3 or 3.0). When pH is higher than 6.0, HCC can remain in monomeric form. However, as the environment changes from acidic to neutral, HCC tetramers break down into dimers. They concluded that the interaction of dimers generates HCC tetrameric forms without a domain-swapping mechanism, as evidenced by the breakdown of tetramers into dimers at pH 7.4 [18].
Another research topic of high interest for studies on PD and AD is related to exosomes. Exosomes play a specific function in the pathophysiology, diagnosis, and prognosis of AD and PD, but their pathophysiological role is not yet fully understood. In a recent review, He et al. discussed the biosynthesis, components, uptake, and functions of exosomes, as well as their pathological roles in AD and PD [19]. In addition to highlighting the use of exosomes as drug carriers for therapies in the central nervous system, the authors provided an overview of recent advances in the clinical implications of exosomes in the treatment and diagnosis of AD and PD.
One of the major challenges in the treatment of neurodegenerative diseases involves finding measures that can facilitate drug penetration through the blood–brain barrier (BBB). The main in vitro BBB models developed thus far for investigating the BBB barrier qualities of the cerebral vasculature are summarized and discussed by Chaulagain et al. They describe multiple existing in vitro models, such as 3D organoid models, microfluidic models, 2D transwell models covering single-layer and co-culture models, and 2D transwell models, together with their construction, permeability measurement, uses, and limitations [20]. Recent achievements and future challenges are discussed and highlighted.
Fasudil, a pan Rho-associated coiled-coil protein kinase (ROCK) inhibitor that dilates blood vessels, has been used for postcerebral stroke. More recently, the use of this medication has been suggested for the treatment of neurodegenerative diseases. Killick et al. conducted a global gene expression analysis in the brains of mice using an AD mouse model, in which fasudil was administered via peripheral intraperitoneal injection [21]. The authors compared the fasudil-induced distinct transcriptional profile in the AD mice model with profiles derived from meta-analyses of other neurodegenerative disorders. The results of their analyses reveal that fasudil stimulates gene expression, in contrast to the gene expression profiles in postmortem brains of patients with neurodegenerative diseases. According to the results of their pathway enrichment analysis, fasudil strongly drives pathways in a reverse manner to disrupted pathways in AD and PD. These findings support the potential repurposed use of fasudil for AD and PD patients, which warrants future investigations.
Numerous lysosomal storage disorders and human neurodegenerative diseases have autophagic dysfunction. This deficiency of lysosome function could exacerbate metabolite buildup and lysosomal distress, subserving the emergence of a neurodegenerative phenotype. Krabbe disease (KD) has recently been linked to abnormalities in the lysosome and autophagy pathways. Large-scale demyelination and dysmyelination are hallmarks of KD, which is triggered by a genetic defect in the lysosomal enzyme galactocerebrosidase (GALC) gene. Galactosylceramide, psychosine, and secondary substrates such as lactosylceramide accumulate due to the loss of function of GALC. Papini et al. used an in vitro cellular starvation model to induce autophagy and observed the cellular reactions and molecular events in patient-isolated fibroblasts [22]. They revealed that the inhibitory AKT-induced beclin-1 phosphorylation and the BCL2–beclin-1 complex jointly contribute to repressing the generation of autophagosomes under starvation [22]. The KD fibroblast model showed autophagic response defects, which could be linked to upregulated AKT activity [22]. These occurrences were not relevant to psychosine accumulation, which was suggested as a key event leading to autophagic dysfunction in KD.
This Special Issue highlights researchers’ continuous investigations of the underlying pathophysiological mechanisms and potential treatments of neurodegenerative diseases. Besides the comprehensive understanding of molecular mechanisms subserving the onset and progression of neurodegenerative diseases, early diagnosis and lifestyle factors such as a healthy diet, exercise, and mental stimulation will also contribute to symptom management and improvement in the life quality of the affected individuals.

Conflicts of Interest

Authors declare no conflict of interest.

References

  1. Zhou, Z.D.; Jankovic, J.; Ashizawa, T.; Tan, E.K. Neurodegenerative diseases associated with non-coding CGG tandem repeat expansions. Nat. Rev. Neurol. 2022, 18, 145–157. [Google Scholar] [PubMed]
  2. Wang, H.; Yang, F.; Zhang, S.; Xin, R.; Sun, Y. Genetic and environmental factors in Alzheimer’s and Parkinson’s diseases and promising therapeutic intervention via fecal microbiota transplantation. NPJ Park. Dis. 2021, 7, 70. [Google Scholar] [CrossRef]
  3. Agnello, L.; Ciaccio, M. Neurodegenerative Diseases: From Molecular Basis to Therapy. Int. J. Mol. Sci. 2022, 23, 12854. [Google Scholar] [CrossRef]
  4. Cannavo, A. Molecular Mechanisms Underlying Chronic and Degenerative Diseases. Int. J. Mol. Sci. 2023, 24, 12507. [Google Scholar] [CrossRef] [PubMed]
  5. Lorenzi, R.M.; Palesi, F.; Castellazzi, G.; Vitali, P.; Anzalone, N.; Bernini, S.; Cotta Ramusino, M.; Sinforiani, E.; Micieli, G.; Costa, A.; et al. Unsuspected Involvement of Spinal Cord in Alzheimer Disease. Front. Cell Neurosci. 2020, 14, 6. [Google Scholar] [CrossRef] [PubMed]
  6. Movio, M.I.; de Lima-Vasconcellos, T.H.; Dos Santos, G.B.; Echeverry, M.B.; Colombo, E.; Mattos, L.S.; Resende, R.R.; Kihara, A.H. Retinal organoids from human-induced pluripotent stem cells: From studying retinal dystrophies to early diagnosis of Alzheimer’s and Parkinson’s disease. Semin. Cell Dev. Biol. 2023, 144, 77–86. [Google Scholar] [CrossRef] [PubMed]
  7. Geng, Z.H.; Zhu, Y.; Li, Q.L.; Zhao, C.; Zhou, P.H. Enteric Nervous System: The Bridge Between the Gut Microbiota and Neurological Disorders. Front. Aging Neurosci. 2022, 14, 810483. [Google Scholar] [CrossRef]
  8. Villa, F.; Carrizzo, A.; Spinelli, C.C.; Ferrario, A.; Malovini, A.; Maciag, A.; Damato, A.; Auricchio, A.; Spinetti, G.; Sangalli, E.; et al. Genetic Analysis Reveals a Longevity-Associated Protein Modulating Endothelial Function and Angiogenesis. Circ. Res. 2015, 117, 333–345. [Google Scholar] [CrossRef]
  9. Di Pardo, A.; Ciaglia, E.; Cattaneo, M.; Maciag, A.; Montella, F.; Lopardo, V.; Ferrario, A.; Villa, F.; Madonna, M.; Amico, E.; et al. The longevity-associated variant of BPIFB4 improves a CXCR4-mediated striatum-microglia crosstalk preventing disease progression in a mouse model of Huntington’s disease. Cell Death Dis. 2020, 11, 546. [Google Scholar] [CrossRef]
  10. Cattaneo, M.; Maciag, A.; Milella, M.S.; Ciaglia, E.; Bruno, A.; Puca, A.A. Longevity-Associated Variant of BPIFB4 Confers Neuroprotection in the STHdh Cell Model of Huntington Disease. Int. J. Mol. Sci. 2022, 23, 15313. [Google Scholar] [CrossRef]
  11. Geng, L.; Fan, L.M.; Liu, F.; Smith, C.; Li, J. Nox2 dependent redox-regulation of microglial response to amyloid-beta stimulation and microgliosis in aging. Sci. Rep. 2020, 10, 1582. [Google Scholar] [CrossRef] [PubMed]
  12. Almeida, C.; Pongilio, R.P.; Movio, M.I.; Higa, G.S.V.; Resende, R.R.; Jiang, J.; Kinjo, E.R.; Kihara, A.H. Distinct Cell-specific Roles of NOX2 and MyD88 in Epileptogenesis. Front. Cell Dev. Biol. 2022, 10, 926776. [Google Scholar] [CrossRef] [PubMed]
  13. Bedard, K.; Krause, K.H. The NOX family of ROS-generating NADPH oxidases: Physiology and pathophysiology. Physiol. Rev. 2007, 87, 245–313. [Google Scholar] [CrossRef]
  14. Benssouina, F.Z.; Parat, F.; Villard, C.; Leloup, L.; Garrouste, F.; Sabatier, J.M.; Ferhat, L.; Kovacic, H. Overexpression of a Novel Noxo1 Mutant Increases Ros Production and Noxo1 Relocalisation. Int. J. Mol. Sci. 2023, 24, 4663. [Google Scholar] [CrossRef] [PubMed]
  15. Ardah, M.T.; Radwan, N.; Khan, E.; Kitada, T.; Haque, M.E. Parkin Precipitates on Mitochondria via Aggregation and Autoubiquitination. Int. J. Mol. Sci. 2023, 24, 9027. [Google Scholar] [CrossRef] [PubMed]
  16. Graves, N.J.; Gambin, Y.; Sierecki, E. alpha-Synuclein Strains and Their Relevance to Parkinson’s Disease, Multiple System Atrophy, and Dementia with Lewy Bodies. Int. J. Mol. Sci. 2023, 24, 12134. [Google Scholar] [CrossRef]
  17. Razzokov, J.; Fazliev, S.; Makhkamov, M.; Marimuthu, P.; Baev, A.; Kurganov, E. Effect of Electric Field on alpha-Synuclein Fibrils: Revealed by Molecular Dynamics Simulations. Int. J. Mol. Sci. 2023, 24, 6312. [Google Scholar] [CrossRef]
  18. Wojciechowska, D.; Taube, M.; Rucinska, K.; Maksim, J.; Kozak, M. Oligomerization of Human Cystatin C-An Amyloidogenic Protein: An Analysis of Small Oligomeric Subspecies. Int. J. Mol. Sci. 2022, 23, 13441. [Google Scholar] [CrossRef]
  19. He, A.; Wang, M.; Li, X.; Chen, H.; Lim, K.; Lu, L.; Zhang, C. Role of Exosomes in the Pathogenesis and Theranostic of Alzheimer’s Disease and Parkinson’s Disease. Int. J. Mol. Sci. 2023, 24, 11054. [Google Scholar] [CrossRef]
  20. Chaulagain, B.; Gothwal, A.; Lamptey, R.N.L.; Trivedi, R.; Mahanta, A.K.; Layek, B.; Singh, J. Experimental Models of In Vitro Blood-Brain Barrier for CNS Drug Delivery: An Evolutionary Perspective. Int. J. Mol. Sci. 2023, 24, 2710. [Google Scholar] [CrossRef]
  21. Killick, R.; Elliott, C.; Ribe, E.; Broadstock, M.; Ballard, C.; Aarsland, D.; Williams, G. Neurodegenerative Disease Associated Pathways in the Brains of Triple Transgenic Alzheimer’s Model Mice Are Reversed Following Two Weeks of Peripheral Administration of Fasudil. Int. J. Mol. Sci. 2023, 24, 11219. [Google Scholar] [CrossRef] [PubMed]
  22. Papini, N.; Todisco, R.; Giussani, P.; Dei Cas, M.; Paroni, R.; Giallanza, C.; Tringali, C. Impaired Autophagy in Krabbe Disease: The Role of BCL2 and Beclin-1 Phosphorylation. Int. J. Mol. Sci. 2023, 24, 5984. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zhou, Z.D.; Kihara, A.H. Neurodegenerative Diseases: Molecular Mechanisms and Therapies. Int. J. Mol. Sci. 2023, 24, 13721. https://doi.org/10.3390/ijms241813721

AMA Style

Zhou ZD, Kihara AH. Neurodegenerative Diseases: Molecular Mechanisms and Therapies. International Journal of Molecular Sciences. 2023; 24(18):13721. https://doi.org/10.3390/ijms241813721

Chicago/Turabian Style

Zhou, Zhi Dong, and Alexandre Hiroaki Kihara. 2023. "Neurodegenerative Diseases: Molecular Mechanisms and Therapies" International Journal of Molecular Sciences 24, no. 18: 13721. https://doi.org/10.3390/ijms241813721

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop