Neurodegeneration, Mitochondria, and Antibiotics: Comparison
Please note this is a comparison between Version 3 by Catherine Yang and Version 2 by Catherine Yang.

Neurodegenerative diseases are characterized by the progressive loss of neurons, synapses, dendrites, and myelin in the central and/or peripheral nervous system. Actual therapeutic options for patients are scarce and merely palliative. Although they affect millions of patients worldwide, the molecular mechanisms underlying these conditions remain unclear. Mitochondrial dysfunction is generally found in neurodegenerative diseases and is believed to be involved in the pathomechanisms of these disorders. Although mitochondrial-targeted treatments are limited, new research findings have unraveled the therapeutic potential of several groups of antibiotics. These drugs possess pleiotropic effects beyond their anti-microbial activity, such as anti-inflammatory or mitochondrial enhancer function. In tThis review we will discuss the ce controversial use of antibiotics as potential therapies in neurodegenerative diseases will be discussed.

  • neurodegeneration
  • mitochondria
  • antibiotics
  • neuroinflammation
  • neurodegenerative diseases

1. Introduction

Neurodegenerative diseases are generally classified according to their clinical presentation, with movement, cognitive, and behavioral disorders being the most common. Diagnosis is generally difficult given the fact that patients normally present heterogeneous clinical features. Very often, neurodegenerative diseases are diagnosed post-mortem via neuropathological evaluation at autopsy. Nevertheless, there is currently great interest among the scientific community in the identification of biomarkers or specific genetic mutations that help clinicians anticipate the onset of these diseases to either treat them when still reversible or slow down their progression [1]. This is especially relevant given the fact that the characteristic protein abnormalities linked to these diseases are present in patients long before the clinical symptoms become noticeable [2][3].

2. Iron Accumulation and Lipid Peroxidation in Neurodegenerative Diseases

Iron accumulation and lipid peroxidation in different areas of the brain have been proposed as key disease-causing factors in many neurodegenerative diseases [4]. Abnormal iron homeostasis generally leads to iron overload, which destroys proteins and lipids via Fenton reactions [5][6]. Excessive iron accumulation and lipid peroxidation are frequently accompanied by oxidative stress, mitochondrial dysfunction, increased lipofuscin granules, and autophagy dysregulation [5][7][8]. Eventually, neuronal cell death occurs by ferroptosis, a cell death process dependent on iron-mediated lipid peroxidation. Minocycline, a second-generation semi-synthetic tetracycline, is a known metal chelator [9], which is the reason why recent studies propose it as a potential treatment for brain iron overload [10]. In mice models, brain non-heme iron and brain iron handling protein levels decreased following minocycline treatment [11]. In fact, absorption of minocycline is significantly decreased by administration with iron supplements [12] and skin hyperpigmentation, a side effect of long-term minocycline therapy, may be related to insoluble minocycline–iron chelation products [13].

3. Neuroinflammation

Neuroinflammation is an inflammatory response within the central nervous system to events that interfere with tissue homeostasis and represents a common denominator in virtually all neurological diseases [14]. Activation of microglia, the main immune effector cells of the brain, contributes to neuronal injury by the release of neurotoxic products [14]. Toll-like receptor 4 (TLR4), expressed on the surface of microglia, plays an important role in mediating lipopolysaccharide (LPS)-induced microglia activation and inflammatory responses. Furthermore, several stress conditions can damage the outer and inner mitochondrial membranes and induce the release of mitochondrial components, such as mitochondrial mtDNA or cardiolipin [15]. These mitochondrial components are recognized as danger-associated molecular patterns (DAMPs), indicating cellular damage and thus eliciting innate immune responses [16]. Detection of both bacterial components, such as LPS, and mitochondrial DAMPs are pro-inflammatory signals. Notably, expression of most of these molecules is not restricted to specialized innate immune cells, such as macrophages, microglia, dendritic cells, or neutrophils, but also occurs in a large number of non-immune cells, including neurons. Antibiotic supplementation has been shown to regulate the neuroinflammatory response reducing its side effects [17][18][19][20].

4. Alzheimer’s Disease

Alzheimer’s disease (AD) is a degenerative disease of the central nervous system with a high incidence in elderly people [21]. The main clinical manifestations are progressive memory loss, cognitive and language communication disorders, and personality changes [22]. Its main pathological features are the appearance of senile plaque (SP) and neurofibrillary tangles (NFTs) in patients’ brains. β-amyloid (Aβ) deposition and abnormally phosphorylated Tau protein deposition are the main components of SP and NFTs, respectively [23]. There is no effective treatment for AD presently available; however, Aβ is now a key established biomarker indicating the development of AD [24]. The aggregation Aβ in AD provokes mitochondrial dysfunction [25] and dynamics impairment, including mitophagy [26].

5. Parkinson’s Disease

Parkinson’s disease (PD) is the second most common degenerative pathology of the central nervous system and its prevalence is higher among people over 65 years old, affecting 1–3% of the population [27][28]. The disease can present a wide range of manifestations. The most recognizable one is a mild tremor that might develop into unilateral resting tremors in the upper limbs over time. However, one out of four PD patients never develops tremors [27]. Other manifestations may involve somatomotor system dysfunction, as well as non-motor symptoms such as neuropsychiatric ones, sleep disorders, or loss of concentration [27][28]. Histopathologically, the presence of neuronal cytoplasmic inclusions called Lewis Bodies (LB), depleted dopamine levels, and loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) are characteristic features of this disease [27][28]. LB are mainly formed by the aggregation of phosphorylated α-synuclein in a misfolded fibrillary stage [27][29][30]. The formation of LB induces neuronal death which affects primarily the dopaminergic neurons in the SNpc [27][31]. The release of α-synuclein to the extracellular compartment might activate microglia and lead to the release of proinflammatory cytokines such as IL-1β through the activation of the NLRP3 inflammasome, thus amplifying neuronal damage [32][33]. Moreover, mitochondrial dysfunction plays a fundamental role in PD [27][28]. Ludtmann et al. suggested that monomeric α-synuclein has a regulatory function in mitochondrial bioenergetics by improving the efficiency of ATP synthase [29][34]. Furthermore, it has been described that aggregated α-synuclein disrupts the vesicular system and, consequently, mitophagy machinery, leading to the accumulation of dysfunctional mitochondria [30][35][36]. Mitochondrial dysfunction enhances ROS production, which may facilitate α-synuclein aggregation and would activate NLRP3 inflammasome [32][37][38]. Synaptic glutamate clearance is also compromised in PD: glutamate transporter-1 (GLT-1) is downregulated in patients causing an imbalance of glutamate homeostasis and excitotoxicity [39][40]. Currently, there is no curative treatment for PD. Most pharmacological formulations are designed to reduce the aggregation of α-synuclein, neuroinflammation, and mitochondrial dysfunction [29][41]. This has led to exploring the efficacy of existing molecules with potential activity against neuroinflammation, α-synuclein aggregation, or mitochondrial dysfunction for their application in PD. In this context, doxycycline has been identified as a potential treatment for PD, since it reduces neuroinflammation and oxidative stress, and prevents the aggregation of α-synuclein in animal models of PD [42][43].

6. Huntington’s Disease

Huntington’s disease (HD) is an autosomal dominant inherited neurodegenerative disease characterized by progressive motor, behavioral, and cognitive decline [44]. The disease results from a CAG trinucleotide repeat expansion in the huntingtin gene (HTT) on chromosome 4. Although its driver gene was discovered in 1993, the pathophysiology of this disease remains mostly unknown [45]. The huntingtin protein is widely expressed and has many functions in human neurons [46] such as intracellular protein trafficking regulation [47], protein scaffolding [48], and synaptic vesicle formation [46][49].

7. Amyotrophic Lateral Sclerosis

Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease that primarily affects the neurons responsible for controlling voluntary muscle movement [50]. ALS affects approximately 4–8 out of 100.000 individuals, with a prognosis for survival of 2 to 5 years. Abundant abnormal protein aggregations have been found in the neurons of CNS in both ALS patients and animal models of ALS [51]. The ubiquitin-proteasome system [52] and the autophagosome-lysosome pathway, including mitophagy, are the most important degradation machinery to clear the aggregated proteins [53]. As seen previously, the balance between protein aggregation and degradation is involved in the pathogenesis of several neurodegenerative diseases [54]. Most ALS-associated identified genes have been functionally implicated in autophagy and/or mitophagy, specifically in the clearance of protein aggregates and/or damaged mitochondria [55]. ALS genes known to function directly in autophagy include OPTN, TBK1, and SQSTM1. Moreover, proteins encoded by the genes C9ORF72, VCP, CHMP2B, VAPB, ALS2, SOD1, and DCTN1 have all been related to vesicular trafficking and may affect autophagy either directly or indirectly [56][57].

8. Prion Diseases

Prion diseases are fatal neurodegenerative disorders with a highly spreading nature. The infectious agent causing prion disease, known as PrPSc, is a pathogenic misfolded and aggregated form of the cellular prion protein, PrPC [58]. Following transmission to a naive host, prions seed the misfolded form of host PrPC in an autocatalytic process, leading to an exponential increase in PrPSc in the brain and the spinal cord that eventually leads to neuronal death [59]. Prions are highly stable and accumulate in the central nervous system from months to years, eventually triggering neurodegeneration and neuronal loss as well as astrocytes and microglia activation [60]. The incubation period of the disease is exceptionally variable and may last from years to weeks. Common symptoms include behavior abnormalities, motor dysfunction, cognitive impairment, and ataxia, depending on the prion type [61]. No therapy is currently available beyond palliative care. Mitochondria of prion-infected animals show morphological and functional abnormalities in the CNS [62]. Alterations in calcium homeostasis related to mitochondria and endoplasmic reticulum dysfunction are typical in prion diseases [63]. Increased calcium concentration promotes mitochondrial membrane loss, enhanced ROS generation, and reduced ATP production, ultimately leading to cellular apoptosis [64]. Taken together these findings suggest that mitochondrial dysfunction may contribute to the neurodegeneration observed in prion diseases. Choi et al. showed that mitochondrial fusion was upregulated in whole brains from prion-infected mice, and that expression levels of mitochondrial fission protein 1 (Fis1) and mitofusin 2 (Mfn2) were elevated in the hippocampus and striatum [65]. Furthermore, the expression of the mitochondrial fission-related protein, Drp1, was significantly reduced in the hippocampus. By inhibiting mitochondrial fission, the mitochondrial network cannot be repaired, which led to a decrease in mitochondrial mass in neurons, most of which were degenerated. These abnormalities were detected in at least four different prion disease mouse models [66]. Despite cellular Drp1 protein levels being decreased in prion-infected neuronal cells both in vitro and in vivo, the levels of the mitochondrial fission protein DLP1 were increased in some prion models. This imbalance results in extensive mitochondrial fragmentation and dysfunction, as well as neuronal death and decreased synaptic plasticity [67].

9. Primary Mitochondrial Diseases

Mitochondrial diseases are a set of highly heterogeneous disorders caused by mutations in either nuclear or mitochondrial genes that primarily affect oxidative phosphorylation and ATP synthesis. These conditions are the most common group of inherited metabolic diseases and one of the most common types of neurological disorders [68][69][70]. In fact, most mitochondrial disease patients present prominent neurologic and myopathic disorders [71]. It is widely known that neurons have a high energy demand and critically depend on mitochondria to maintain synaptic transmission through the regulation of ATP and calcium levels [72]. Early onset mitochondrial diseases are severe clinical entities often caused by autosomal recessive mutations in nuclear genes (nDNA). Examples of these include Leigh syndrome, caused by mutations in the mitochondrial oxidative phosphorylation (OXPHOS) complexes and their assembly factors [73], and Alpers–Huttenlocher syndrome, due to nDNA mutations in the mtDNA polymerase gene [74]. Other high-energy demanding tissues such as the skeletal and cardiac muscles can also be affected in patients with these diseases.
Mitochondrial diseases diagnosed in adulthood affect around 1 in 4300 adults, and are mostly caused by mtDNA mutations (approx. 87% of cases) [75]. Mitochondrial syndromes caused by mtDNA mutations that affect the nervous system include Leber’s hereditary optic neuropathy (LHON), myoclonic epilepsy with red-ragged fibers (MERRF), and mitochondrial encephalopathy, lactic acidosis, and stroke-like episodes (MELAS) [76].
The activation of mitochondrial compensatory mechanisms has been associated with the regulation of mitophagy and mitochondrial biogenesis as well as with a beneficial mild induction of ROS signaling pathways. Animals presenting impaired cardiac mitophagy and a consequent accumulation of damaged ROS-overproducing mitochondria develop cardiomyopathy. It has been observed that this condition can be improved through ROS-dependent activation of mitophagy, which can act as a mitochondrial quality control mechanism to prevent vicious cycles of ROS formation and mitochondrial dysfunction [77]. Furthermore, muscle-specific knockout mice for COX15, a complex IV assembly protein, are able to express alternative oxidases (AOX) that bypass respiratory complexes III and IV, thus transferring electrons directly to oxygen. In doing so, they exhibit decreased ROS generation, PGC-1α signaling activation, and increased lifespan [78]. Livers from adult mice depleted from superoxide dismutase 2 during embryonic development display mitochondrial adaptive responses with increased mitochondrial biogenesis and antioxidant defenses and decreased ROS levels [79]. Taken together, these results show the impressive ability of mitochondria to adapt to cellular insults.
There is no proven treatment for most primary mitochondrial diseases, only palliative therapies. Triggering mitohormesis with antibiotics such as tetracyclines or their derivatives could open the door to new therapeutic perspectives for these severe pathologies.

10. Cerebral Ischemia

Cerebral ischemia (CI) and its implications are currently one of the leading causes of mortality and morbidity worldwide. Given that the brain is one of the most energy-consuming organs, disruption of the blood supply, and thus of nutrient and oxygen availability, can result in severe neuronal damage [80]. There is increasing evidence that acute neuronal damage induced by cerebral ischemia promotes protein aggregation, suggesting a connection between the pathomechanisms of neurodegenerative diseases and stroke [81][82][83]. Thus, it seems that brain ischemia contributes to the development of Alzheimer’s disease-like neurodegeneration through various mechanisms, including accumulation of amyloid protein precursor, tau protein phosphorylation, neuroinflammation, dysregulation of Alzheimer-related genes, neuronal cell death, synaptic dysfunction, white matter alterations, and brain atrophy [84][85].

11. Neuropsychiatric Diseases: Schizophrenia and Depression

This section is included since major psychiatric and neurodegenerative diseases share genetic susceptibility and pathophysiology [86]. Thus, many causal proteins and interacting proteins, and the central role of synaptic transmission, immune and mitochondrial function are processes that participate in the shared pathogenesis. Schizophrenia is a chronic neuropsychiatric disease that affects around 1% of the world population [87]. It may present with a wide range of symptoms either classified as positive (delusion, hallucination, and paranoia) or negative (amotivation, anhedonia, avolition, asociality, and flat affect). Negative and positive symptoms are caused by different alterations in neurotransmitter functions. Positive symptoms are caused by hyperfunction in the mesolimbic dopamine pathway [88], whereas negative symptoms may include abnormal GABAα receptors, disruption of glutamatergic systems, and abnormal microglial activation [87]. Pathophysiologically, neuroinflammation plays a key role in the disease, as evidenced by the high microglial activity that has been found in these patients [89][90]. This altered function of the brain immune system has been related to the dysfunction of glutamatergic and dopaminergic neurotransmissions [91], as well as to gray matter volume loss [90]. On the other hand, energy metabolism is also altered in the brain of patients with schizophrenia. This alteration may be due to mitochondrial dysfunction increased lactate levels and acidemia found in patients’ brains [92]. This alteration may also affect neurotransmission, since dopamine levels and NMDA receptor activity are highly sensitive to pH changes [92]. Furthermore, an elevated synthesis and release of dopamine in presynaptic neurons has been found, suggesting that presynaptic mitochondria are exposed to higher levels of dopamine, a complex I inhibitor. Thus, it generates a decrease in ATP synthesis without increasing oxidative stress [88][92]. Furthermore, there are alterations in the expression of genes related to energy metabolism in the brain of schizophrenia patients [92]. Although there is no cure for schizophrenia, it is usually treated with dopamine receptor antagonists [88] which control positive symptoms but have little to no effect on negative symptoms [92][93][94].

12. Conclusions

As presented across this research, the aetiology of neurodegenerative diseases is extremely complex and can be etiologically diverse. However, most of these diseases share the common feature of mitochondrial dysfunction. The application of antibiotics to treat mitochondrial insults has always been controversial for plenty and reasonable reasons . However, not all antibiotics function in the same way or at the same dosage. Thanks to their pleiotropic effects, these drugs may open new possibilities for the treatment of neurodegenerative disease beyond their antimicrobial activity. However, more research is needed to address the potential side effects of their chronic administration such as the risk of dissemination of antibiotic-resistant pathogenic strains. For this reason, the development of antibiotics derivatives without antimicrobial activity but retaining their neuroprotective properties will be another interesting research field in the future.

References

  1. Beach, T.G. A Review of Biomarkers for Neurodegenerative Disease: Will They Swing Us Across the Valley? Neurol. Ther. 2017, 6 (Suppl. S1), 5–13.
  2. Evidente, V.G.; Adler, C.H.; Sabbagh, M.N.; Connor, D.J.; Hentz, J.G.; Caviness, J.N.; Sue, L.I.; Beach, T.G. Neuropathological findings of PSP in the elderly without clinical PSP: Possible incidental PSP? Park. Relat. Disord. 2011, 17, 365–371.
  3. Frigerio, R.; Fujishiro, H.; Ahn, T.B.; Josephs, K.A.; Maraganore, D.M.; DelleDonne, A.; Parisi, J.E.; Klos, K.J.; Boeve, B.F.; Dickson, D.W.; et al. Incidental Lewy body disease: Do some cases represent a preclinical stage of dementia with Lewy bodies? Neurobiol. Aging 2011, 32, 857–863.
  4. Stockwell, B.R.; Angeli, J.P.F.; Bayir, H.; Bush, A.I.; Conrad, M.; Dixon, S.J.; Fulda, S.; Gascón, S.; Hatzios, S.K.; Kagan, V.E.; et al. Ferroptosis: A Regulated Cell Death Nexus Linking Metabolism, Redox Biology, and Disease. Cell 2017, 171, 273–285.
  5. Ke, Y.; Qian, Z.M. Brain iron metabolism: Neurobiology and neurochemistry. Prog. Neurobiol. 2007, 83, 149–173.
  6. Li, C.; Wu, Z.; Xue, H.; Gao, Q.; Zhang, Y.; Wang, C.; Zhao, P. Ferroptosis contributes to hypoxic-ischemic brain injury in neonatal rats: Role of the SIRT1/Nrf2/GPx4 signaling pathway. CNS Neurosci. Ther. 2022, 28, 2268–2280.
  7. Álvarez-Córdoba, M.; Khoury, A.F.; Villanueva-Paz, M.; Gómez-Navarro, C.; Villalón-García, I.; Suárez-Rivero, J.M.; Povea-Cabello, S.; de la Mata, M.; Cotán, D.; Talaverón-Rey, M.; et al. Pantothenate Rescues Iron Accumulation in Pantothenate Kinase-Associated Neurodegeneration Depending on the Type of Mutation. Mol. Neurobiol. 2018, 56, 3638–3656.
  8. Corti, O.; Blomgren, K.; Poletti, A.; Beart, P.M. Autophagy in neurodegeneration: New insights underpinning therapy for neurological diseases. J. Neurochem. 2020, 154, 354–371.
  9. Grenier, D.; Huot, M.P.; Mayrand, D. Iron-chelating activity of tetracyclines and its impact on the susceptibility of Actinobacillus actinomycetemcomitans to these antibiotics. Antimicrob. Agents Chemother. 2000, 44, 763–766.
  10. Zhao, F.; Xi, G.; Liu, W.; Keep, R.F.; Hua, Y. Minocycline Attenuates Iron-Induced Brain Injury. Acta Neurochir. Suppl. 2016, 121, 361–365.
  11. Zhao, F.; Hua, Y.; He, Y.; Keep, R.; Xi, G. Minocycline-Induced Attenuation of Iron Overload and Brain Injury after Experimental Intracerebral Hemorrhage. Stroke 2011, 42, 3587–3593.
  12. Leyden, J.J. Absorption of minocycline hydrochloride and tetracycline hydrochloride: Effect of food, milk, and iron. J. Am. Acad. Dermatol. 1985, 12, 308–312.
  13. Geria, A.N.; Tajirian, A.L.; Kihiczak, G.; Schwartz, R.A. Minocycline-induced skin pigmentation: An update. Acta Dermatovenerol. Croat. 2009, 17, 123–126.
  14. Leng, F.; Edison, P. Neuroinflammation and microglial activation in Alzheimer disease: Where do we go from here? Nat. Rev. Neurol. 2021, 17, 157–172.
  15. Kolmychkova, K.I.; Zhelankin, A.V.; Karagodin, V.P.; Orekhov, A.N. Mitochondria and inflammation. Zhurnal Patol. Fiziol. Eksperimentalnaia Ter. 2016, 60, 114–121.
  16. Banoth, B.; Cassel, S.L. Mitochondria in innate immune signaling. Transl. Res. 2018, 202, 52–68.
  17. Acuña, L.; Hamadat, S.; Corbalán, N.S.; González-Lizárraga, F.; dos-Santos-Pereira, M.; Rocca, J.; Sepúlveda Díaz, J.; Del-Bel, E.; Papy-García, D.; Chehín, R.N.; et al. Rifampicin and Its Derivative Rifampicin Quinone Reduce Microglial Inflammatory Responses and Neuro-degeneration Induced In Vitro by alpha-Synuclein Fibrillary Aggregates. Cells 2019, 8, 776.
  18. Cankaya, S.; Cankaya, B.; Kilic, U.; Kilic, E.; Yulug, B. The therapeutic role of minocycline in Parkinson’s disease. Drugs Context 2019, 8, 212553.
  19. Zusso, M.; Lunardi, V.; Franceschini, D.; Pagetta, A.; Lo, R.; Stifani, S.; Frigo, A.C.; Giusti, P.; Moro, S. Ciprofloxacin and levofloxacin attenuate microglia inflammatory response via TLR4/NF-kB pathway. J. Neuroinflammation 2019, 16, 148.
  20. Khanna, D.; Kalra, S. Minocycline and Doxycycline: More Than Antibiotics. Curr. Mol. Pharmacol. 2021, 14, 1046–1065.
  21. Nelson, P.T.; Braak, H.; Markesbery, W.R. Neuropathology and Cognitive Impairment in Alzheimer Disease: A Complex but Coherent Relationship. J. Neuropathol. Exp. Neurol. 2009, 68, 1–14.
  22. Alexiou, A.; Kamal, M.A.; Ashraf, G.M. Editorial: The Alzheimer’s Disease Challenge. Front. Neurosci. 2019, 13, 768.
  23. Martins, R.N.; Villemagne, V.; Sohrabi, H.R.; Chatterjee, P.; Shah, T.M.; Verdile, G.; Fraser, P.; Taddei, K.; Gupta, V.B.; Rainey-Smith, S.R.; et al. Alzheimer’s Disease: A Journey from Amyloid Peptides and Oxidative Stress, to Biomarker Technologies and Disease Prevention Strategies-Gains from AIBL and DIAN Cohort Studies. J. Alzheimers. Dis. 2018, 62, 965–992.
  24. Morris, G.P.; Clark, I.A.; Vissel, B. Questions concerning the role of amyloid-beta in the definition, aetiology and diagnosis of Alzheimer’s disease. Acta Neuropathol. 2018, 136, 663–689.
  25. Swerdlow, R.H. Mitochondria and Mitochondrial Cascades in Alzheimer’s Disease. J. Alzheimer’s Dis. 2018, 62, 1403–1416.
  26. Schmukler, E.; Solomon, S.; Simonovitch, S.; Goldshmit, Y.; Wolfson, E.; Michaelson, D.M.; Pinkas-Kramarski, R. Altered mitochondrial dynamics and function in APOE4-expressing astrocytes. Cell Death Dis. 2020, 11, 578.
  27. Raza, C.; Anjum, R.; Shakeel, N.U.A. Parkinson’s disease: Mechanisms, translational models and management strategies. Life Sci. 2019, 226, 77–90.
  28. Grünewald, A.; Kumar, K.R.; Sue, C.M. New insights into the complex role of mitochondria in Parkinson’s disease. Prog. Neurobiol. 2018, 177, 73–93.
  29. Socias, S.B.; González-Lizárraga, F.; Avila, C.L.; Vera, C.; Acuña, L.; Sepulveda-Diaz, J.E.; Del-Bel, E.; Raisman-Vozari, R.; Chehin, R.N. Exploiting the therapeutic potential of ready-to-use drugs: Repurposing antibiotics against amyloid aggregation in neurodegenerative diseases. Prog. Neurobiol. 2018, 162, 17–36.
  30. Dominguez-Meijide, A.; Parrales, V.; Vasili, E.; González-Lizárraga, F.; König, A.; Lázaro, D.F.; Lannuzel, A.; Haik, S.; Del Bel, E.; Chehín, R.; et al. Doxycycline inhibits α-synuclein-associated pathologies in vitro and in vivo. Neurobiol. Dis. 2021, 151, 105256.
  31. Volpicelli-Daley, L.A.; Luk, K.C.; Patel, T.P.; Tanik, S.A.; Riddle, D.M.; Stieber, A.; Meaney, D.F.; Trojanowski, J.Q.; Lee, V.M.-Y. Exogenous α-Synuclein Fibrils Induce Lewy Body Pathology Leading to Synaptic Dysfunction and Neuron Death. Neuron 2011, 72, 57–71.
  32. Bortolanza, M.; Nascimento, G.C.; Socias, S.B.; Ploper, D.; Chehín, R.N.; Raisman-Vozari, R.; Del-Bel, E. Tetracycline repurposing in neurodegeneration: Focus on Parkinson’s disease. J. Neural Transm. 2018, 125, 1403–1415.
  33. Codolo, G.; Plotegher, N.; Pozzobon, T.; Brucale, M.; Tessari, I.; Bubacco, L.; de Bernard, M. Triggering of inflammasome by aggregated alpha-synuclein, an inflammatory response in synucleinopathies. PLoS ONE 2013, 8, e55375.
  34. Ludtmann, M.H.; Angelova, P.R.; Ninkina, N.N.; Gandhi, S.; Buchman, V.L.; Abramov, A.Y. Monomeric Alpha-Synuclein Exerts a Physiological Role on Brain ATP Synthase. J. Neurosci. 2016, 36, 10510–10521.
  35. Lindström, V.; Gustafsson, G.; Sanders, L.H.; Howlett, E.H.; Sigvardson, J.; Kasrayan, A.; Ingelsson, M.; Bergström, J.; Erlandsson, A. Extensive uptake of α-synuclein oligomers in astrocytes results in sustained intracellular deposits and mitochondrial damage. Mol. Cell. Neurosci. 2017, 82, 143–156.
  36. Liang, B.; Tamm, L.K. Solution NMR of SNAREs, complexin and alpha-synuclein in association with membrane-mimetics. Prog. Nucl. Magn. Reason. Spectrosc. 2018, 105, 41–53.
  37. Liang, Y.; Zhou, T.; Chen, Y.; Lin, D.; Jing, X.; Peng, S.; Zheng, D.; Zeng, Z.; Lei, M.; Wu, X.; et al. Rifampicin inhibits rotenone-induced microglial inflammation via enhancement of autophagy. Neurotoxicology 2017, 63, 137–145.
  38. Yurtsever, İ.; Üstündağ, Ü.V.; Ünal, İ.; Ateş, P.S.; Emekli-Alturfan, E. Rifampicin decreases neuroinflammation to maintain mitochondrial function and calcium homeostasis in rote-none-treated zebrafish. Drug Chem. Toxicol. 2022, 45, 1544–1551.
  39. Chotibut, T.; Meadows, S.; Kasanga, E.A.; McInnis, T.; Cantu, M.A.; Bishop, C.; Salvatore, M.F. Ceftriaxone reduces L-dopa-induced dyskinesia severity in 6-hydroxydopamine Parkinson’s disease model. Mov. Disord. 2017, 32, 1547–1556.
  40. Kaur, B.; Prakash, A. Ceftriaxone attenuates glutamate-mediated neuro-inflammation and restores BDNF in MPTP model of Parkinson’s disease in rats. Pathophysiology 2017, 24, 71–79.
  41. Han, J.; Kim, S.J.; Ryu, M.J.; Jang, Y.; Lee, M.J.; Ju, X.; Lee, Y.L.; Cui, J.; Shong, M.; Heo, J.Y.; et al. Chloramphenicol Mitigates Oxidative Stress by Inhibiting Translation of Mitochondrial Complex I in Dopaminergic Neurons of Toxin-Induced Parkinson’s Disease Model. Oxidative Med. Cell. Longev. 2019, 2019, 4174803.
  42. González-Lizárraga, F.; Socías, S.B.; Ávila, C.L.; Torres-Bugeau, C.M.; Barbosa, L.R.S.; Binolfi, A.; Sepúlveda-Díaz, J.E.; Del-Bel, E.; Fernandez, C.O.; Papy-Garcia, D.; et al. Repurposing doxycycline for synucleinopathies: Remodelling of α-synuclein oligomers towards non-toxic parallel beta-sheet structured species. Sci. Rep. 2017, 7, srep41755.
  43. Amaral, L.D.; dos Santos, N.A.G.; Sisti, F.M.; Del Bel, E.; dos Santos, A.C. The antibiotic doxycycline mimics the NGF signaling in PC12 cells: A relevant mechanism for neuroprotection. Chem. Interact. 2021, 341, 109454.
  44. Wyant, K.J.; Ridder, A.J.; Dayalu, P. Huntington’s Disease-Update on Treatments. Curr. Neurol. Neurosci. Rep. 2017, 17, 33.
  45. A novel gene containing a trinucleotide repeat that is expanded and unstable on Huntington’s disease chromosomes. The Hunting-ton’s Disease Collaborative Research Group. Cell 1993, 72, 971–983.
  46. Saudou, F.; Humbert, S. The Biology of Huntingtin. Neuron 2016, 89, 910–926.
  47. Cornett, J.; Cao, F.; Wang, C.-E.; Ross, C.A.; Bates, G.P.; Li, S.-H.; Li, X.-J. Polyglutamine expansion of huntingtin impairs its nuclear export. Nat. Genet. 2005, 37, 198–204.
  48. Godin, J.D.; Colombo, K.; Molina-Calavita, M.; Keryer, G.; Zala, D.; Charrin, B.C.; Dietrich, P.; Volvert, M.-L.; Guillemot, F.; Dragatsis, I.; et al. Huntingtin Is Required for Mitotic Spindle Orientation and Mammalian Neurogenesis. Neuron 2010, 67, 392–406.
  49. McKinstry, S.U.; Karadeniz, Y.B.; Worthington, A.K.; Hayrapetyan, V.Y.; Ozlu, M.I.; Serafin-Molina, K.; Risher, W.C.; Ustunkaya, T.; Dragatsis, I.; Zeitlin, S.; et al. Huntingtin Is Required for Normal Excitatory Synapse Development in Cortical and Striatal Circuits. J. Neurosci. 2014, 34, 9455–9472.
  50. Hulisz, D. Amyotrophic lateral sclerosis: Disease state overview. Am. J. Manag. Care 2018, 24, S320–S326.
  51. Rumfeldt, J.A.; Lepock, J.R.; Meiering, E.M. Unfolding and Folding Kinetics of Amyotrophic Lateral Sclerosis-Associated Mutant Cu, Zn Superoxide Dismutases. J. Mol. Biol. 2009, 385, 278–298.
  52. Nandi, D.; Tahiliani, P.; Kumar, A.; Chandu, D. The ubiquitin-proteasome system. J. Biosci. 2006, 31, 137–155.
  53. Evans, C.S.; Holzbaur, E.L. Autophagy and mitophagy in ALS. Neurobiol. Dis. 2018, 122, 35–40.
  54. Rubinsztein, D.C. The roles of intracellular protein-degradation pathways in neurodegeneration. Nature 2006, 443, 780–786.
  55. Taylor, J.P.; Brown, R.H., Jr.; Cleveland, D.W. Decoding ALS: From genes to mechanism. Nature 2016, 539, 197–206.
  56. Chua, J.P.; De Calbiac, H.; Kabashi, E.; Barmada, S.J. Autophagy and ALS: Mechanistic insights and therapeutic implications. Autophagy 2021, 18, 254–282.
  57. Yang, C.; Zhang, X. Research progress on vesicular trafficking in amyotrophic lateral sclerosis. J. Zhejiang Univ. (Medical Sci.) 2022, 51, 380–387.
  58. Prusiner, S. Novel proteinaceous infectious particles cause scrapie. Science 1982, 216, 136–144.
  59. Prusiner, S.B.; Scott, M.R.; DeArmond, S.J.; Cohen, F.E. Prion Protein Biology. Cell 1998, 93, 337–348.
  60. DeArmond, S.J.; Prusiner, S.B. Etiology and pathogenesis of prion diseases. Am. J. Pathol. 1995, 146, 785–811.
  61. Takada, L.T.; Geschwind, M.D. Prion diseases. Semin. Neurol. 2013, 33, 348–356.
  62. Park, J.H.; Kim, B.H.; Park, S.J.; Jin, J.K.; Jeon, Y.C.; Wen, G.Y.; Shin, H.Y.; Carp, R.I.; Kim, Y.S. Association of endothelial nitric oxide synthase and mitochondrial dysfunction in the hippocampus of scrapie-infected mice. Hippocampus 2011, 21, 319–333.
  63. Moon, J.-H.; Park, S.-Y. Prion peptide-mediated calcium level alteration governs neuronal cell damage through AMPK-autophagy flux. Cell Commun. Signal. 2020, 18, 109.
  64. Ferreiro, E.; Oliveira, C.R.; Pereira, C.M. The release of calcium from the endoplasmic reticulum induced by amyloid-beta and prion peptides activates the mitochondrial apoptotic pathway. Neurobiol. Dis. 2008, 30, 331–342.
  65. Choi, H.-S.; Choi, Y.-G.; Shin, H.-Y.; Oh, J.-M.; Park, J.-H.; Kim, J.-I.; Carp, R.I.; Choi, E.-K.; Kim, Y.-S. Dysfunction of mitochondrial dynamics in the brains of scrapie-infected mice. Biochem. Biophys. Res. Commun. 2014, 448, 157–162.
  66. Yang, X.-D.; Shi, Q.; Sun, J.; Lv, Y.; Ma, Y.; Chen, C.; Xiao, K.; Zhou, W.; Dong, X.-P. Aberrant Alterations of Mitochondrial Factors Drp1 and Opa1 in the Brains of Scrapie Experiment Rodents. J. Mol. Neurosci. 2016, 61, 368–378.
  67. Li, C.; Wang, D.; Wu, W.; Yang, W.; Shah, S.Z.A.; Zhao, Y.; Duan, Y.; Wang, L.; Zhou, X.; Zhao, D.; et al. DLP1-dependent mitochondrial fragmentation and redistribution mediate prion-associated mitochondrial dysfunction and neuronal death. Aging Cell 2017, 17, e12693.
  68. Burtscher, J.; Romani, M.; Bernardo, G.; Popa, T.; Ziviani, E.; Hummel, F.C.; Sorrentino, V.; Millet, G.P. Boosting mitochondrial health to counteract neurodegeneration. Prog. Neurobiol. 2022, 215, 102289.
  69. Gorman, G.S.; Chinnery, P.F.; DiMauro, S.; Hirano, M.; Koga, Y.; McFarland, R.; Suomalainen, A.; Thorburn, D.R.; Zeviani, M.; Turnbull, D.M. Mitochondrial diseases. Nat. Rev. Dis. Primers 2016, 2, 16080.
  70. Zeviani, M.; Viscomi, C. Mitochondrial Neurodegeneration. Cells 2022, 11, 637.
  71. Nelson, I.; Hanna, M.G.; Alsanjari, N.; Scaravilli, F.; Morgan-Hughes, J.A.; Harding, A.E. A new mitochondrial DNA mutation associated with progressive dementia and chorea: A clinical, pathological, and molecular genetic study. Ann. Neurol. 1995, 37, 400–403.
  72. Chen, H.; Chan, D.C. Critical dependence of neurons on mitochondrial dynamics. Curr. Opin. Cell Biol. 2006, 18, 453–459.
  73. Lake, N.J.; Bird, M.J.; Isohanni, P.; Paetau, A. Leigh syndrome: Neuropathology and pathogenesis. J. Neuropathol. Exp. Neurol. 2015, 74, 482–492.
  74. Harding, B.N. Review Article: Progressive Neuronal Degeneration of Childhood with Liver Disease (Alpers-Huttenlocher Syndrome): A Personal Review. J. Child Neurol. 1990, 5, 273–287.
  75. Gorman, G.S.; Schaefer, A.M.; Ng, Y.; Gomez, N.; Blakely, E.L.; Alston, C.L.; Feeney, C.; Horvath, R.; Yu-Wai-Man, P.; Chinnery, P.F.; et al. Prevalence of nuclear and mitochondrial DNA mutations related to adult mitochondrial disease. Ann. Neurol. 2015, 77, 753–759.
  76. Russell, O.; Turnbull, D. Mitochondrial DNA disease—Molecular insights and potential routes to a cure. Exp. Cell Res. 2014, 325, 38–43.
  77. Song, M.; Chen, Y.; Gong, G.; Murphy, E.; Rabinovitch, P.S.; Dorn, G.W. Super-Suppression of Mitochondrial Reactive Oxygen Species Signaling Impairs Compensatory Autophagy in Primary Mitophagic Cardiomyopathy. Circ. Res. 2014, 115, 348–353.
  78. Dogan, S.A.; Cerutti, R.; Benincá, C.; Brea-Calvo, G.; Jacobs, H.T.; Zeviani, M.; Szibor, M.; Viscomi, C. Perturbed Redox Signaling Exacerbates a Mitochondrial Myopathy. Cell Metab. 2018, 28, 764–775.e5.
  79. Cox, C.S.; McKay, S.E.; Holmbeck, M.A.; Christian, B.E.; Scortea, A.C.; Tsay, A.J.; Newman, L.E.; Shadel, G.S. Mitohormesis in Mice via Sustained Basal Activation of Mitochondrial and Antioxidant Signaling. Cell Metab. 2018, 28, 776–786.e5.
  80. Lin, H.W.; Lee, R.H.C.; Lee, M.H.H.; Wu, C.Y.C.; Silva, A.C.; Possoit, H.E.; Hsieh, T.-H.; Minagar, A. Cerebral ischemia and neuroregeneration. Neural Regen. Res. 2018, 13, 373–385.
  81. Hayashi, T.; Takada, K.; Matsuda, M. Post-transient ischemia increase in ubiquitin conjugates in the early reperfusion. Neuroreport 1992, 3, 519–520.
  82. Kahl, A.; Blanco, I.; Jackman, K.; Baskar, J.; Milaganur Mohan, H.; Rodney-Sandy, R.; Zhang, S.; Iadecola, C.; Hochrainer, K. Cerebral ischemia induces the aggregation of proteins linked to neurodegenerative diseases. Sci. Rep. 2018, 8, 2701.
  83. Yang, W.; Sheng, H.; Warner, D.S.; Paschen, W. Transient Focal Cerebral Ischemia Induces a Dramatic Activation of Small Ubiquitin-Like Modifier Conjugation. J. Cereb. Blood Flow Metab. 2008, 28, 892–896.
  84. Pluta, R. Brain ischemia as a bridge to Alzheimer’s disease. Neural Regen. Res. 2022, 17, 791.
  85. Pluta, R.; Januszewski, S.; Czuczwar, S.J. Brain Ischemia as a Prelude to Alzheimer’s Disease. Front. Aging Neurosci. 2021, 13, 636653.
  86. Wingo, T.S.; Liu, Y.; Gerasimov, E.S.; Vattathil, S.M.; Wynne, M.E.; Liu, J.; Lori, A.; Faundez, V.; Bennett, D.A.; Seyfried, N.T.; et al. Shared mechanisms across the major psychiatric and neurodegenerative diseases. Nat. Commun. 2022, 13, 4314.
  87. Zhang, L.; Zheng, H.; Wu, R.; Zhu, F.; Kosten, T.R.; Zhang, X.-Y.; Zhao, J. Minocycline adjunctive treatment to risperidone for negative symptoms in schizophrenia: Association with pro-inflammatory cytokine levels. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2018, 85, 69–76.
  88. Ben-Shachar, D. The bimodal mechanism of interaction between dopamine and mitochondria as reflected in Parkinson’s disease and in schizophrenia. J. Neural Transm. 2019, 127, 159–168.
  89. Inta, D.; Lang, U.E.; Borgwardt, S.; Meyer-Lindenberg, A.; Gass, P. Microglia Activation and Schizophrenia: Lessons from the Effects of Minocycline on Postnatal Neurogenesis, Neuronal Survival and Synaptic Pruning. Schizophr. Bull. 2016, 43, 493–496.
  90. Krynicki, C.R.; Dazzan, P.; Pariante, C.M.; Barnes, N.M.; Vincent, R.C.; Roberts, A.; Giordano, A.; Watson, A.; Suckling, J.; Barnes, T.R.; et al. Deconstructing depression and negative symptoms of schizophrenia; differential and longitudinal immune correlates, and response to minocycline treatment. Brain Behav. Immun. 2021, 91, 498–504.
  91. Çakici, N.; Van Beveren, N.J.M.; Judge-Hundal, G.; Koola, M.M.; Sommer, I.E.C. An update on the efficacy of anti-inflammatory agents for patients with schizophrenia: A meta-analysis. Psychol. Med. 2019, 49, 2307–2319.
  92. Park, H.-J.; Choi, I.; Leem, K.-H. Decreased Brain pH and Pathophysiology in Schizophrenia. Int. J. Mol. Sci. 2021, 22, 8358.
  93. Roberts, R.C. Mitochondrial dysfunction in schizophrenia: With a focus on postmortem studies. Mitochondrion 2020, 56, 91–101.
  94. Kelly, D.L.; Sullivan, K.M.; McEvoy, J.P.; McMahon, R.P.; Wehring, H.J.; Gold, J.M.; Liu, F.; Warfel, D.; Vyas, G.; Richardson, C.M.; et al. Adjunctive Minocycline in Clozapine-Treated Schizophrenia Patients with Persistent Symptoms. J. Clin. Psychopharmacol. 2015, 35, 374–381.
More
Video Production Service