Kynurenines in Neurodegenerative Diseases: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Contributor: , ,

Neurodegenerative diseases are multifactorial, initiated by a series of the causative complex which develops into a certain clinical picture. The pathogenesis and disease course vary from patient to patient. Thus, it should be likewise to the treatment. Peripheral biomarkers are to play a central role for tailoring a personalized therapeutic plan for patients who suffered from neurodegenerative diseases such as Alzheimer’s disease, Parkinson’s disease, and multiple sclerosis, among others. Nevertheless, the use of biomarkers in clinical practice is still underappreciated and data presented in biomarker research for clinical use is still uncompelling, compared to the abundant data available for drug research and development. So is the case with kynurenines (KYNs) and the kynurenine pathway (KP) enzymes, which have been associated with a wide range of diseases including cancer, autoimmune diseases, inflammatory diseases, neurologic diseases, and psychiatric disorders. 

  • neurodegenerative disease
  • Alzheimer’s disease
  • Parkinson’s disease
  • amyotrophic lateral sclerosis
  • Huntington’s disease
  • multiple sclerosis
  • stroke
  • tryptophan
  • kynurenine

1. Neurodegenerative Diseases

1.1. Alzheimer’s Disease

Alzheimer’s disease (AD) is the most common chronic neurodegenerative disease with an insidious onset of progressive cognitive deteriorations, particularly memory impairment. Motor or sensory dysfunctions are not prominent in the early stage. Motor and autonomic dysfunctions are associated with the comorbidities such as Parkinson’s disease (PD) with dementia, dementia with Lewy bodies, or vascular dementia [1]. Anxiety is common, besides apathy, depression, aggression, or sleep disorder [2]. Cortical atrophy of the frontal, temporal, and parietal lobes, enlargement of the temporal horn of the lateral ventricle, and atrophy of the entorhinal cortex, amygdala, and hippocampus are pathognomonic findings in patients with AD [3]. Abnormal deposit of insoluble proteinaceous material amyloid beta (Aβ) in the neuron and glial cells is mainly located in the atrophic lesions of AD patients [4]. Tau protein aggregates are associated with Aβ deposits, but it is considered secondary to amyloidosis [5]. Disturbance of calcium homeostasis was observed, and calcium-related proteins were proposed to be diagnostic and therapeutic biomarkers in AD [6].
The serum levels of TRP, KYNA, 3-HK, QUIN, and PIC were measured. The levels of 3-HK were significantly increased in the serum of patients with AD compared to those with major depression or with cognitive impairments. 3-HK is permeable to the BBB in contrast to a downstream metabolite QUIN and may be associated with higher levels of QUIN in the brain of AD patients [7]. An urgent need for biomarkers for the detection of the early stage AD was declared to expedite the early intervention by disease-modifying agents [7].
3-HK and other KP intermediates are possible candidates of early stage biomarkers [7]. The roles of the KP in the pathogenesis of AD were described [8][9][10][11][12]. QUIN was found localized with hyperphosphorylated tau in the cortical neurons of the brain of AD patients and to induce the phosphorylation of tau in human brains [9]. AD patients who have higher QUIN levels performed worse on the CAMCOG (the cognitive and self-contained part of the Cambridge Examination for Mental Disorders of the Elderly) test, suggesting the levels QUIN are associated with the cognitive impairment level [13] (Table 1). QUIN is a strong oxidant, and the presence of oxidative stress was reported in AD, involving mitochondria dysfunction, microRNA, and microRNA-gene interaction [14][15].
Table 1. Changes of metabolites of the kynurenine pathway in neurologic diseases. ↑: increase, ↓: decrease, *: tendency, not statistically significant.
Increased IDO-1 activity was associated with reduced cognitive performance, while IDO-1 inhibitor coptisine decreased the activation of microglia and astrocytes, prevented neuron loss, reduced Aβ plaque formation, and ameliorated impaired cognition in A b PP/PS1 mice [45][46]. KMO inhibitor JM6 prevented spatial memory deficits, anxiety related behavior, and synaptic loss in APP-Tg mice [43]. Furthermore, IDO is associated with the senile plaques [10]. Finally, the increased levels of KYNA were specific to cerebrospinal fluid (CSF) in AD, compared to that of frontotemporal dementia (FTD) and ALS [32] (Table 2).
Table 2. Changes of metabolites of the kynurenine pathway in neurologic diseases. ↑: increase, ↓: decrease.

1.2. Parkinson’s Disease

PD is a progressive neurodegenerative disorder that predominantly affects motor functions including muscle rigidity, tremors, and changes in speech and gait. Main motor dysfunctions are bradykinesia, resting tremors, and rigidity which are largely due to the dopaminergic nigrostriatal denervation in the early stages of PD. However, psychobehavioral symptoms including psychosis, hallucinations, depression, and anxiety are not rare, which are present before the motor complaints [50][52]. Neurodegeneration and gliosis of the pars compacta of the substantia nigra (SNpc) and the presence of Lewy bodies (LBs) in pigment nuclei are pathognomonic of PD [53]. LBs contain the abnormal aggregates of misfolded alpha-synuclein (α-syn). Accumulation of aggregated α-syn in oligodendrocytes forms glial cytoplasmic inclusions. The mechanisms that govern α-syn fibrillization and LB formation in the brain remain poorly understood [54].
Alterations of TRP metabolism, glutamate excitotoxicity, and the gut-brain-axis have been shown associated with the pathogenesis of PD [55][56]. The identification of a risk marker is of particular interest because most of the dopaminergic neuros in the SNpc is not functioning at the time of the diagnosis [57]. KYNA levels and KYNA/KYN ratios were found significantly lower, while the levels of QUIN and ratios of QUIN/KYNA were observed significantly higher in the plasma of PD patients compared to healthy controls [23][58]. Those patients who were in advanced stage, Hoehn-Yahr stage more than 2, showed lower levels of KYNA and ratios of KYNA/KYN, while higher levels of QUIN and ratios of QUIN/KYNA compared to PD patients in early stage, Hoehn-Yahr stage ≤2 and healthy controls [58]. Moreover, receiver operating characteristic curve analysis suggested a QUIN/KYNA ratio as a potential biomarker for PD with good sensitivity and specificity. Stratified analysis showed that changes of the KYN pathway metabolites were more characteristic in PD patients in advanced stage [58]. Altered KYN metabolism and KYNA levels were reported in the brain samples of PD patients. The levels of KYNA were lower in the frontal cortex, putamen, and SNpc, while the levels of 3-HK were higher in the putamen, frontal cortex, SNpc, and CSF of PD patients [23][29]. The elevated 3-HK levels in CSF evidenced a possible excitotoxic disease mechanism in PD and 3-HK as a potential predictive biomarker [23] (Table 1).
Single nucleotide polymorphisms (SNP) of IDO-1 rs7820268 and rs9657182 were found associated with the late onset of PD [59]. The activities of KAT II and levels of KYNA were increased in the red blood cells; however, the activities of KAT I and KAT II were lower and the levels of KYNA tended to be lower in the plasma of PD patients [41] (Table 2). A systematic review reported the increased levels of neurotoxic KYNs and the decreased levels of neuroprotective KYNs in general, suggesting a significant shift toward the production of QUIN in the KP in PD [1]. Alteration of the KP is a distinguished characteristic in PD and may contribute to the pathogenesis of PD. Highly active retrotranposition competent LINE-1s was linked to the risk and progression of PD. making it a possible risk and therapeutic biomarkers [60]. Thus, the identification of PD-specific biomarkers in the blood, CSF, stool, or urine sample may make it possible to reveal the pathogenesis, make an early stage diagnosis, observe the disease progression, and monitor therapeutic effects.

1.3. Amyotrophic Lateral Sclerosis

Amyotrophic lateral sclerosis (ALS) is a group of progressive neurodegenerative disease which mainly affects neurons controlling voluntary muscles. ALS often presents fasciculation, myasthenia, or dysarthria initially. It involves the muscles responsible to move, speak, eat, and breathe in later stage [61]. ALS patients present a wide range of mild symptoms including autonomic, GI, cardiovascular, and neuropsychiatric manifestations including depression and anxiety [62][63][64]. The most common genetic mutations are the GGGGCC expansion in C9ORF72, present in approximately 30–47% of familial ALS cases and the SOD 1 mutations [65][66]. This hexanucleotide expansion is most often accompanied with the presence of cytoplasmic inclusions containing transactive response DNA-binding protein of 43 kDa (TDP-43) [67]. TDP-43 proteinopathy is characteristic to ALS [65]. TDP-43 is found in the lower motor neurons in the spinal cord and brainstem and the upper motor neurons in the motor cortex. In the late stage of ALS and ALS patients with dementia, TDP-43 can be found in the hippocampus, amygdala, and cortex [4]. New mutations were found in the genes of chromosome 9 open reading frame 72 (C9orf72), SOD 1, and senataxin in Hungarian ALS patients [68][69]. SNP of the vitamin D receptor gene rs7975232 (ApaI) was found associated with ALS [70]. Besides genetic predisposition, the pathogenesis of ALS is associated with ROS, mitochondrial dysfunction, intracellular calcium aggregation, and protein aggregation, glutamate excitotoxicity, and autoimmune inflammatory process [70][71][72][73].
The levels of KYNA was observed higher in the brain of patients with bulbar onset of ALS, compared to healthy control or patients with limb onset [36]. Moreover, the levels of KYNA were higher in CSF of patients with severe clinical status, compared to healthy controls [36]. Meanwhile lower levels of KYNA were detected in the serum of patients with severe clinical status, compared to healthy controls and patients with mild clinical status [36] (Table 1). Therefore, the serum level of KYNA possibly indicates the severity of the disease and can be a potential prognostic biomarker. The neuronal and microglial expression of IDO were elevated and the levels of QUIN were higher in the motor cortex and spinal cord of ALS patients [36] (Table 2). The levels of TRP, KYN, and QUIN were elevated in CSF of ALS patients [36]. The levels of TRP, KYN, and QUIN were elevated, and the level of PIC was decreased in the serum of ALS patients [36]. Furthermore, the ratios of 3-HK/XA were decreased in the serum of ALS patients compared to patients with FTD [17] (Table 1). The median survivaltime ranges from 20 to 48 months, but only 10–20% of ALS patients survive longer than ten years with worse prognosis in older age and bulbar onset. There is no option for the treatment of ALS. Thus, a search for predictive and therapeutic biomarkers are of particular interest.

1.4. Huntington’s Disease

Huntington’s disease (HD) is an autosomal-dominant neurodegenerative disease with progressive and irreversible motor dysfunctions, leading to coordination problem, gait difficulties, cognitive dysfunction, and behavioral changes. Mild autonomic symptoms including orthostatic hypotension, excessive perspiration, and tachycardia are present in mild HD, while vegetative symptoms are prominent in the advanced stages [71]. Pathological findings in HD are degeneration and neural loss of the striatum, especially the caudate nuclei which innervate the cerebral cortex, pallidum, thalamus, brainstem, and cerebellum. The pathological changes correlate with disability. In the cerebellum, thalamus, and brain stem, abundant ballooned neurons were observed. Abnormal huntingtin proteins are associated with ballooning cell death which ruptures the membrane to swell like a balloon [71].
The activation of the neurotoxic branch of the KP is verified in the CNS. The levels of 3-HK and QUIN were elevated and the activity of 3-HAO was increased in the striatum where the loss of the nerve cell is the most prominent [24][50]. The levels of KYNA and the activity of KAT were decreased in the brain [37]. Toxoplasma gondii infection elevated the IDO activity in the brain and resulted significantly earlier death of the transgenic mouse model of HD compared to the HD mice without infection and the wild type, suggesting that the IDO activation accelerated the disease progression [72]. Lower TRP, higher KYN levels, and higher KYN/TRP ratios were observed in the serum of HD patients, suggesting the presence of higher IDO activity [18]. The levels of KYNA, the activity of KAT, and the levels of 3-HK and 3-HAA were all decreased in plasma [36]. The inflammatory status was well correlated with the levels of AA and the levels of TRP were negatively correlated with the severity of symptoms and the number of CAG repeats [19]. AA levels may be a good biomarker to indicate the inflammatory status in HD (Table 1).

1.5. Multiple Sclerosis

Multiple sclerosis (MS) is an autoimmune demyelinating neurodegenerative disease. Common symptoms of MS range widely from motor dysfunction, autonomic symptoms to psychobehavioral manifestations including gait difficulties, paresthesia, vision problems, vertigo, incontinence, sexual problems, pain, cognitive dysfunctions, emotional changes, and depression [73]. Numerous glial scars, called plaques which develop in the white matter and spinal cord are pathognomonic to MS [74].
Disturbance of the KP and altered levels of KYN metabolites in MS patients were reported previously [25][40][75][76]. The levels of TRP were significantly lower in the serum and CSF samples of MS patients, suggesting the activation of the KP metabolism [20].The lower TRP level may be used as a potential biomarker in the screening of MS [19][21]. Proinflammatory cytokines including IFNs and TNF-α activated IDO-1 expression, resulting in the activation of the KP [77]. However, IDO-1 inhibition upon the disease induction significantly exacerbated the disease severity in the experimental autoimmune encephalitis (EAE) model of MS in mouse [78]. It was probably caused by the increased pool of available TRP which facilitated T cell proliferation. The activation and expression of IDO may become a useful biomarker to monitor the clinical course of relapsing-remitting multiple sclerosis (RRMS) and a predictive biomarker for the development of flares of MS. Moreover, therapeutic modulation of IDO activity may be beneficial in MS [40]. An imbalance of neurotoxic and neuroprotective KYN metabolites was considered involved in the pathogenesis of MS [25][41][79]. The activities of KAT I and KAT II enzymes were significantly higher in the red blood cells of MS patients compared to healthy control and the higher KAT activities were correlated with increased KYNA levels in the plasma of MS patients, suggesting the possible induction of neuroprotection against excitoneurotoxicity [44][47]. Furthermore, the levels of KYNA were increased in the CSF of MS patients during acute phase, while the KYNA levels were decreased in the inactive chronic phase of MS [25][26]. Monitoring the redox status including proteins, lipids, and nucleic acids together with the KP components was proposed to build a personalized treatment plan for MS patients [80] (Table 1).
The activity and expression of KMO and the level of 3-HK were increased and KMO inhibitor Ro61- 8048 decreased the QUIN concentration in the spinal cord of EAE model of MS in rats [41]. A Ro61- 8048 prodrug KM6 significantly increased KYNA levels in mouse models of HD and AD [43]. The activity of KMO may be responsible for deviating from the KYNA branch towards the production of downstream neurotoxic metabolites. The activation of IDO-1 inhibits T cell activation, which appears beneficial to MS, but it can lead to the increased production of neurotoxic KYNs, eventually contributing to the progression of MS [43][81]. The KP metabolites were proposed to be potential prognostic and predictive biomarkers for MS. The levels KYNA and PIC were increased in RRMS, but not in secondary progressive multiple sclerosis (SPMS) or primary progressive (PPMS) and the levels of 3-HK and QUIN were increased in both SPMS and PPMS [69]. The QUIN/KYNA ratios were moderately correlated with the severity of MS [75] (Table 1). Alteration of TRP metabolism is more relevant to the pathogenesis of MS than inflammation and a profile of the serum KP metabolites may be a suitable predictive biomarker for MS. Especially, the QUIN/KYNA ratio may become a useful predictive biomarker for neurodegeneration [75]. In general, the levels of KP metabolites can be suitable biomarkers for diagnosis of MS subtypes, monitoring the severity, and/or identifying therapeutic targets.

2. Other Relevant Diseases

Potential use of KYNs as biomarkers are under extensive research not only for neurologic diseases, but also psychiatric disorders. KYN and KYNA were found to be predictive biomarkers for the treatment of escitalopram in depression. KYNA is both a diagnostic and a predictive biomarker for depression as well [82]. 3-HK and KYNA were proposed to be prognostic biomarkers of depression and disability in poststroke patients [83]. Other KP metabolites such as XA and CA were rarely documented. An extremely low concentration of CA was reported to have anti-psychotic activities in mice and the levels of CA was reduced in the prefrontal cortex in patients with schizophrenia [84]. The glutamatergic nervous system was proposed to be a therapeutic biomarker for mood disorders including depression [85]. Furthermore, a simultaneous intervention in the NMDA receptor and α7nAchR was suggested by novel combination for the treatment of schizophrenia [86]. Longitudinal plasma samples were studied in search of a certain plasma protein profiles as a predictive biomarker for the treatment of depression [87]. Therapeutic biomarkers are under rigorous search for depression, anxiety, and dementia through endogenous neuropeptides, agonists, and their synthetic analogues [88][89][90][91]. Omega-3 polyunsaturated fatty acids which bind G protein-coupled receptor GPR120 in the GPR120 signaling pathway was proposed to be a therapeutic biomarker for the treatment of schizophrenia [92]. The treatment of metabolic dysfunction by nutraceuticals in ageing and neurodegenerative diseases was proposed [93]. Biomarkers are not only limited to molecules, but can also be any measurable indicators for risk, diagnosis, prognosis, disease course, and therapeutic targets. Depression was reported a risk factor for AD and dementia, and dyslipidemia treatment reduced this risk in patients with diabetes mellitus. Thus, depression is a risk biomarker and preventable in patients with dyslipidemia [94]. The presence of depression after acute stroke and transient ischemic attack increased mortality and disability within 12 months, suggesting depression as a prognostic biomarker in cerebral ischemia [95]. Depression and anxiety can be treatable by psychedelic psilocybin in patients with terminal illness [96]. Interestingly, depression is a single psychobehavioral component of dementia, which can be ameliorated by animal-assisted and pet-robot interventions in dementia patients [97]. Depression is indeed a therapeutic biomarker.

This entry is adapted from the peer-reviewed paper 10.3390/ijms21249338

References

  1. Tanaka, M.; Toldi, J.; Vécsei, L. Exploring the Etiological Links behind Neurodegenerative Diseases: Inflammatory Cytokines and Bioactive Kynurenines. Int. J. Mol. Sci. 2020, 21, 2431.
  2. Zhao, Q.F.; Tan, L.; Wang, H.F.; Jiang, T.; Tan, M.S.; Tan, L.; Xu, W.; Li, J.Q.; Wang, J.; Lai, T.J.; et al. The prevalence of neuropsychiatric symptoms in Alzheimer’s disease: Systematic review and meta-analysis. J. Affect. Disord. 2016, 190, 264–271.
  3. DeTure, M.A.; Dickson, D.W. The neuropathological diagnosis of Alzheimer’s disease. Mol. Neurodegener. 2019, 14, 32.
  4. Dugger, B.N.; Dickson, D.W. Pathology of Neurodegenerative Diseases. Cold Spring Harb. Perspect. Biol. 2017, 9, a028035.
  5. Ambar Akkaoui, M.; Geoffroy, P.A.; Roze, E.; Degos, B.; Garcin, B. Functional Motor Symptoms in Parkinson’s Disease and Functional Parkinsonism: A Systematic Review. J. Neuropsychiatry Clin. Neurosci. 2020, 32, 4–13.
  6. O’Day, D.H. Calmodulin Binding Proteins and Alzheimer’s Disease: Biomarkers, Regulatory Enzymes and Receptors That Are Regulated by Calmodulin. Int. J. Mol. Sci. 2020, 21, 7344.
  7. Schwarz, M.J.; Guillemin, G.J.; Teipel, S.J.; Buerger, K.; Hampel, H. Increased 3-hydroxykynurenine serum concentrations differentiate Alzheimer’s disease patients from controls. Eur. Arch. Psychiatry Clin. Neurosci. 2013, 26, 345–352.
  8. Guillemin, G.J.; Brew, B.J.; Noonan, C.E.; Takikawa, O.; Cullen, K.M. Indoleamine 2,3 dioxygenase and quinolinic acid immunoreactivity in Alzheimer’s disease hippocampus. Neuropathol. Appl. Neurobiol. 2005, 31, 395–404.
  9. Rahman, A.; Ting, K.; Cullen, K.M.; Braidy, N.; Brew, B.J.; Guillemin, G.J. The excitotoxin quinolinic acid induces tau phosphorylation in human neurons. PLoS ONE 2009, 4, e6344.
  10. Bonda, D.J.; Mailankot, M.; Stone, J.G.; Garrett, M.R.; Staniszewska, M.; Castellani, R.J.; Siedlak, S.L.; Zhu, X.; Lee, H.G.; Perry, G.; et al. Indoleamine 2,3-dioxygenase and 3-hydroxykynurenine modifications are found in the neuropathology of Alzheimer’s disease. Redox Rep. 2010, 15, 161–168.
  11. Wu, W.; Nicolazzo, J.A.; Wen, L.; Chung, R.; Stankovic, R.; Bao, S.S.; Lim, C.K.; Brew, B.J.; Cullen, K.M.; Guillemin, G.J. Expression of tryptophan 2,3-dioxygenase and production of kynurenine pathway metabolites in triple transgenic mice and human Alzheimer’s disease brain. PLoS ONE 2013, 8, e59749.
  12. van der Velpen, V.; Teav, T.; Gallart-Ayala, H.; Mehl, F.; Konz, I.; Clark, C.; Oikonomidi, A.; Peyratout, G.; Henry, H.; Delorenzi, M.; et al. Systemic and central nervous system metabolic alterations in Alzheimer’s disease. Alzheimers Res. Ther. 2019, 11, 93.
  13. Giil, L.M.; Midttun, O.; Refsum, H.; Ulvik, A.; Advani, R.; Smith, A.D.; Ueland, P.M. Kynurenine Pathway Metabolites in Alzheimer’s Disease. J. Alzheimers Dis. 2017, 60, 495–504.
  14. Catanesi, M.; d’Angelo, M.; Tupone, M.G.; Benedetti, E.; Giordano, A.; Castelli, V.; Cimini, A. MicroRNAs Dysregulation and Mitochondrial Dysfunction in Neurodegenerative Diseases. Int. J. Mol. Sci. 2020, 21, 5986.
  15. Brito, L.M.; Ribeiro-dos-Santos, Â.; Vidal, A.F.; de Araújo, G.S. on behalf of the Alzheimer’s Disease Neuroimaging Initiative. Differential Expression and miRNA–Gene Interactions in Early and Late Mild Cognitive Impairment. Biology 2020, 9, 251.
  16. Gulaj, E.; Pawlak, K.; Bien, B.; Pawlak, D. Kynurenine and its metabolites in Alzheimer’s disease patients. Adv. Med. Sci. 2010, 55, 204–211.
  17. Chen, Y.; Stankovic, R.; Cullen, K.M.; Meininger, V.; Garner, B.; Coggan, S.; Grant, R.; Brew, B.J.; Guillemin, G.J. The kynurenine pathway and inflammation in amyotrophic lateral sclerosis. Neurotox. Res. 2010, 18, 132–142.
  18. Widner, B.; Leblhuber, F.; Walli, J.; Tilz, G.P.; Demel, U.; Fuchs, D. Degradation of tryptophan in neurodegenerative disorders. Adv. Exp. Med. Biol. 1999, 467, 133–138.
  19. Forrest, C.M.; Mackay, G.M.; Stoy, N.; Spiden, S.L.; Taylor, R.; Stone, T.W.; Darlington, L.G. Blood levels of kynurenines, interleukin-23 and soluble human leucocyte antigen-G at different stages of Huntington’s disease. J. Neurochem. 2010, 112, 112–122.
  20. Monaco, F.; Fumero, S.; Mondino, A.; Mutani, R. Plasma and cerebrospinal fluid tryptophan in multiple sclerosis and degenerative diseases. J. Neurol. Neurosurg. Psychiatry 1979, 42, 640–641.
  21. Cocco, E.; Murgia, F.; Lorefice, L.; Barberini, L.; Poddighe, S.; Frau, J.; Fenu, G.; Coghe, G.; Murru, M.R.; Murru, R.; et al. (1)H-NMR analysis provides a metabolomic profile of patients with multiple sclerosis. Neurol. Neuroimmunol. Neuroinflamm. 2016, 3, e185.
  22. Heilman, P.L.; Wang, E.W.; Lewis, M.M.; Krzyzanowski, S.; Capan, C.D.; Burmeister, A.R.; Du, G.; Escobar Galvis, M.L.; Brundin, P.; Huang, X.; et al. Tryptophan Metabolites Are Associated With Symptoms and Nigral Pathology in Parkinson’s Disease. Mov. Disord. 2020.
  23. Chang, K.H.; Cheng, M.L.; Tang, H.Y.; Huang, C.Y.; Wu, Y.R.; Chen, C.M. Alternations of Metabolic Profile and Kynurenine Metabolism in the Plasma of Parkinson’s Disease. Mol. Neurobiol. 2018, 55, 6319–6328.
  24. Guidetti, P.; Luthi-Carter, R.E.; Augood, S.J.; Schwarcz, R. Neostriatal and cortical quinolinate levels are increased in early grade Huntington’s disease. Neurobiol. Dis. 2004, 17, 455–461.
  25. Rejdak, K.; Bartosik-Psujek, H.; Dobosz, B.; Kocki, T.; Grieb, P.; Giovannoni, G.; Turski, W.A.; Stelmasiak, Z. Decreased level of kynurenic acid in cerebrospinal fluid of relapsing-onset multiple sclerosis patients. Neurosci. Lett. 2002, 331, 63–65.
  26. Rejdak, K.; Petzold, A.; Kocki, T.; Kurzepa, J.; Grieb, P.; Turski, W.A.; Stelmasiak, Z. Astrocytic activation in relation to inflammatory markers during clinical exacerbation of relapsing-remitting multiple sclerosis. J. Neural Transm. (Vienna) 2007, 114, 1011–1015.
  27. Chatterjee, P.; Goozee, K.; Lim, C.K.; James, I.; Shen, K.; Jacobs, K.R.; Sohrabi, H.R.; Shah, T.; Asih, P.R.; Dave, P.; et al. Alterations in serum kynurenine pathway metabolites in individuals with high neocortical amyloid-beta load: A pilot study. Sci. Rep. 2018, 8, 8008.
  28. Ogawa, T.; Matson, W.R.; Beal, M.F.; Myers, R.H.; Bird, E.D.; Milbury, P.; Saso, S. Kynurenine pathway abnormalities in Parkinson’s disease. Neurology 1992, 42, 1702–1706.
  29. Lewitt, P.A.; Li, J.; Lu, M.; Beach, T.G.; Adler, C.H.; Guo, L.; Arizona Parkinson’s Disease Consortium. 3-hydroxykynurenine and other Parkinson’s disease biomarkers discovered by metabolomic analysis. Mov. Disord. 2013, 28, 1653–1660.
  30. Pearson, S.J.; Reynolds, G.P. Increased brain concentrations of a neurotoxin, 3-hydroxykynurenine, in Huntington’s disease. Neurosci. Lett. 1992, 144, 199–201.
  31. Stoy, N.; Mackay, G.M.; Forrest, C.M.; Christofides, J.; Egerton, M.; Stone, T.W.; Darlington, L.G. Tryptophan metabolism and oxidative stress in patients with Huntington’s disease. J. Neurochem. 2005, 93, 611–623.
  32. González-Sánchez, M.; Jiménez, J.; Narváez, A.; Antequera, D.; Llamas-Velasco, S.; Martín, A.-S.; Molina Arjona, J.A.; López de Munain, A.; Lleó Bisa, A.; Marco, M.-P.; et al. Kynurenic Acid Levels are Increased in the CSF of Alzheimer’s Disease Patients. Biomolecules 2020, 10, 571.
  33. Heyes, M.P.; Saito, K.; Crowley, J.S.; Davis, L.E.; Demitrack, M.A.; Der, M.; Dilling, L.A.; Elia, J.; Kruesi, M.J.; Lackner, A.; et al. Quinolinic acid and kynurenine pathway metabolism in inflammatory and non-inflammatory neurological disease. Brain 1992, 115 Pt 5, 1249–1273.
  34. Baran, H.; Jellinger, K.; Deecke, L. Kynurenine metabolism in Alzheimer’s disease. J. Neural Transm. (Vienna) 1999, 106, 165–181.
  35. Hartai, Z.; Juhasz, A.; Rimanoczy, A.; Janaky, T.; Donko, T.; Dux, L.; Penke, B.; Toth, G.K.; Janka, Z.; Kalman, J. Decreased serum and red blood cell kynurenic acid levels in Alzheimer’s disease. Neurochem. Int. 2007, 50, 308–313.
  36. Ilzecka, J.; Kocki, T.; Stelmasiak, Z.; Turski, W.A. Endogenous protectant kynurenic acid in amyotrophic lateral sclerosis. Acta Neurol. Scand. 2003, 107, 412–418.
  37. Beal, M.F.; Matson, W.R.; Swartz, K.J.; Gamache, P.H.; Bird, E.D. Kynurenine pathway measurements in Huntington’s disease striatum: Evidence for reduced formation of kynurenic acid. J. Neurochem. 1990, 55, 1327–1339.
  38. Beal, M.F.; Matson, W.R.; Storey, E.; Milbury, P.; Ryan, E.A.; Ogawa, T.; Bird, E.D. Kynurenic acid concentrations are reduced in Huntington’s disease cerebral cortex. J. Neurol. Sci. 1992, 108, 80–87.
  39. Jauch, D.; Urbanska, E.M.; Guidetti, P.; Bird, E.D.; Vonsattel, J.P.; Whetsell, W.O., Jr.; Schwarcz, R. Dysfunction of brain kynurenic acid metabolism in Huntington’s disease: Focus on kynurenine aminotransferases. J. Neurol. Sci. 1995, 130, 39–47.
  40. Mancuso, R.; Hernis, A.; Agostini, S.; Rovaris, M.; Caputo, D.; Fuchs, D.; Clerici, M. Indoleamine 2,3 Dioxygenase (IDO) Expression and Activity in Relapsing-Remitting Multiple Sclerosis. PLoS ONE 2015, 10, e0130715.
  41. Chiarugi, A.; Cozzi, A.; Ballerini, C.; Massacesi, L.; Moroni, F. Kynurenine 3-mono-oxygenase activity and neurotoxic kynurenine metabolites increase in the spinal cord of rats with experimental allergic encephalomyelitis. Neuroscience 2001, 102, 687–695.
  42. Hartai, Z.; Klivenyi, P.; Janaky, T.; Penke, B.; Dux, L.; Vécsei, L. Kynurenine metabolism in plasma and in red blood cells in Parkinson’s disease. J. Neurol. Sci. 2005, 239, 31–35.
  43. Zwilling, D.; Huang, S.Y.; Sathyasaikumar, K.V.; Notarangelo, F.M.; Guidetti, P.; Wu, H.Q.; Lee, J.; Truong, J.; Andrews-Zwilling, Y.; Hsieh, E.; et al. Kynurenine 3-monooxygenase inhibition in blood ameliorates neurodegeneration. Cell 2011, 145, 863–874.
  44. Bai, J.H.; Zheng, Y.L.; Yu, Y.P. Urinary kynurenine as a biomarker for Parkinson’s disease. Neurol. Sci. 2020.
  45. Widner, B.; Leblhuber, F.; Walli, J.; Tilz, G.P.; Demel, U.; Fuchs, D. Tryptophan degradation and immune activation in Alzheimer’s disease. J. Neural Transm. (Vienna) 2000, 107, 343–353.
  46. Yu, D.; Tao, B.B.; Yang, Y.Y.; Du, L.S.; Yang, S.S.; He, X.J.; Zhu, Y.W.; Yan, J.K.; Yang, Q. The IDO inhibitor coptisine ameliorates cognitive impairment in a mouse model of Alzheimer’s disease. J. Alzheimers Dis. 2015, 43, 291–302.
  47. Widner, B.; Leblhuber, F.; Fuchs, D. Increased neopterin production and tryptophan degradation in advanced Parkinson’s disease. J. Neural Transm. (Vienna) 2002, 109, 181–189.
  48. Knyihar-Csillik, E.; Csillik, B.; Pakaski, M.; Krisztin-Peva, B.; Dobo, E.; Okuno, E.; Vecsei, L. Decreased expression of kynurenine aminotransferase-I (KAT-I) in the substantia nigra of mice after 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) treatment. Neuroscience 2004, 126, 899–914.
  49. Hartai, Z.; Klivenyi, P.; Janaky, T.; Penke, B.; Dux, L.; Vecsei, L. Kynurenine metabolism in multiple sclerosis. Acta Neurol. Scand. 2005, 112, 93–96.
  50. Jacobs, K.R.; Lim, C.K.; Blennow, K.; Zetterberg, H.; Chatterjee, P.; Martins, R.N.; Brew, B.J.; Guillemin, G.J.; Lovejoy, D.B. Correlation between plasma and CSF concentrations of kynurenine pathway metabolites in Alzheimer’s disease and relationship to amyloid-beta and tau. Neurobiol. Aging 2019, 80, 11–20.
  51. Schwarcz, R.; Okuno, E.; White, R.J.; Bird, E.D.; Whetsell, W.O., Jr. 3-Hydroxyanthranilate oxygenase activity is increased in the brains of Huntington disease victims. Proc. Natl. Acad. Sci. USA 1988, 85, 4079–4081.
  52. Papagno, C.; Trojano, L. Cognitive and behavioral disorders in Parkinson’s disease: An update. I: Cognitive impairments. Neurol. Sci. 2018, 39, 215–223.
  53. Broen, M.P.; Narayen, N.E.; Kuijf, M.L.; Dissanayaka, N.N.; Leentjens, A.F. Prevalence of anxiety in Parkinson’s disease: A systematic review and meta-analysis. Mov. Disord. 2016, 31, 1125–1133.
  54. Török, N.; Majlath, Z.; Szalardy, L.; Vécsei, L. Investigational alpha-synuclein aggregation inhibitors: Hope for Parkinson’s disease. Expert Opin. Investig. Drugs 2016, 25, 1281–1294.
  55. Caputi, V.; Giron, M.C. Microbiome-Gut-Brain Axis and Toll-Like Receptors in Parkinson’s Disease. Int. J. Mol. Sci. 2018, 19, 1689.
  56. Mahul-Mellier, A.L.; Burtscher, J.; Maharjan, N.; Weerens, L.; Croisier, M.; Kuttler, F.; Leleu, M.; Knott, G.W.; Lashuel, H.A. The process of Lewy body formation, rather than simply α-synuclein fibrillization, is one of the major drivers of neurodegeneration. Proc. Natl. Acad. Sci. USA 2020, 117, 4971–4982.
  57. Török, N.; Török, R.; Szolnoki, Z.; Somogyvari, F.; Klivenyi, P.; Vécsei, L. The Genetic Link between Parkinson’s Disease and the Kynurenine Pathway Is Still Missing. Parkinsons Dis. 2015, 2015, 474135.
  58. Dal Ben, M.; Bongiovanni, R.; Tuniz, S.; Fioriti, E.; Tiribelli, C.; Moretti, R.; Gazzin, S. Earliest Mechanisms of Dopaminergic Neurons Sufferance in a Novel Slow Progressing Ex Vivo Model of Parkinson Disease in Rat Organotypic Cultures of Substantia Nigra. Int. J. Mol. Sci. 2019, 20, 2224.
  59. Török, N.; Maszlag-Török, R.; Molnár, K.; Szolnoki, Z.; Somogyvári, F.; Boda, K.; Tanaka, M.; Klivényi, P.; Vécsei, L. Single Nucleotide Polymorphisms of Indoleamine 2,3-Dioxygenase 1 Influenced the Age Onset of Parkinson’s Disease. Preprints 2020.
  60. Pfaff, A.L.; Bubb, V.J.; Quinn, J.P.; Koks, S. An Increased Burden of Highly Active Retrotransposition Competent L1s Is Associated with Parkinson’s Disease Risk and Progression in the PPMI Cohort. Int. J. Mol. Sci. 2020, 21, 6562.
  61. Rojas, P.; Ramirez, A.I.; Fernandez-Albarral, J.A.; Lopez-Cuenca, I.; Salobrar-Garcia, E.; Cadena, M.; Elvira-Hurtado, L.; Salazar, J.J.; de Hoz, R.; Ramirez, J.M. Amyotrophic Lateral Sclerosis: A Neurodegenerative Motor Neuron Disease with Ocular Involvement. Front. Neurosci. 2020, 14, 566858.
  62. Piccione, E.A.; Sletten, D.M.; Staff, N.P.; Low, P.A. Autonomic system and amyotrophic lateral sclerosis. Muscle Nerve 2015, 51, 676–979.
  63. Fang, T.; Jozsa, F.; Al-Chalabi, A. Nonmotor Symptoms in Amyotrophic Lateral Sclerosis: A Systematic Review. Int. Rev. Neurobiol. 2017, 134, 1409–1441.
  64. Carvalho, T.L.; de Almeida, L.M.; Lorega, C.M.; Barata, M.F.; Ferreira, M.L.; de Brito-Marques, P.R.; Correia Cda, C. Depression and anxiety in individuals with amyotrophic lateral sclerosis: A systematic review. Trends Psychiatry Psychother. 2016, 38, 1–5.
  65. Renton, A.E.; Majounie, E.; Waite, A.; Simon-Sanchez, J.; Rollinson, S.; Gibbs, J.R.; Schymick, J.C.; Laaksovirta, H.; van Swieten, J.C.; Myllykangas, L.; et al. A hexanucleotide repeat expansion in C9ORF72 is the cause of chromosome 9p21-linked ALS-FTD. Neuron 2011, 72, 257–268.
  66. Zou, Z.Y.; Zhou, Z.R.; Che, C.H.; Liu, C.Y.; He, R.L.; Huang, H.P. Genetic epidemiology of amyotrophic lateral sclerosis: A systematic review and meta-analysis. J. Neurol. Neurosurg. Psychiatry 2017, 88, 540–549.
  67. Neumann, M.; Sampathu, D.M.; Kwong, L.K.; Truax, A.C.; Micsenyi, M.C.; Chou, T.T.; Bruce, J.; Schuck, T.; Grossman, M.; Clark, C.M.; et al. Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science 2006, 314, 130–133.
  68. Tripolszki, K.; Csanyi, B.; Nagy, D.; Ratti, A.; Tiloca, C.; Silani, V.; Kereszty, E.; Török, N.; Vecsei, L.; Engelhardt, J.I.; et al. Genetic analysis of the SOD1 and C9ORF72 genes in Hungarian patients with amyotrophic lateral sclerosis. Neurobiol. Aging 2017, 53, 195.e1–195.e5.
  69. Tripolszki, K.; Török, D.; Goudenege, D.; Farkas, K.; Sulak, A.; Török, N.; Engelhardt, J.I.; Klivényi, P.; Procaccio, V.; Nagy, N.; et al. High-throughput sequencing revealed a novel SETX mutation in a Hungarian patient with amyotrophic lateral sclerosis. Brain Behav. 2017, 7, e00669.
  70. Török, N.; Török, R.; Klivényi, P.; Engelhardt, J.; Vécsei, L. Investigation of vitamin D receptor polymorphisms in amyotrophic lateral sclerosis. Acta Neurol. Scand. 2016, 133, 302–308.
  71. Dash, D.; Mestre, T.A. Therapeutic Update on Huntington’s Disease: Symptomatic Treatments and Emerging Disease-Modifying Therapies. Neurotherapeutics 2020.
  72. Donley, D.W.; Olson, A.R.; Raisbeck, M.F.; Fox, J.H.; Gigley, J.P. Huntingtons Disease Mice Infected with Toxoplasma gondii Demonstrate Early Kynurenine Pathway Activation, Altered CD8+ T-Cell Responses, and Premature Mortality. PLoS ONE 2016, 11, e0162404.
  73. Boeschoten, R.E.; Braamse, A.M.J.; Beekman, A.T.F.; Cuijpers, P.; van Oppen, P.; Dekker, J.; Uitdehaag, B.M.J. Prevalence of depression and anxiety in Multiple Sclerosis: A systematic review and meta-analysis. J. Neurol. Sci. 2017, 372, 331–341.
  74. Huang, W.J.; Chen, W.W.; Zhang, X. Multiple sclerosis: Pathology, diagnosis and treatments. Exp. Ther. Med. 2017, 13, 3163–3166.
  75. Lim, C.K.; Bilgin, A.; Lovejoy, D.B.; Tan, V.; Bustamante, S.; Taylor, B.V.; Bessede, A.; Brew, B.J.; Guillemin, G.J. Kynurenine pathway metabolomics predicts and provides mechanistic insight into multiple sclerosis progression. Sci. Rep. 2017, 7, 41473.
  76. Platten, M.; Ho, P.P.; Youssef, S.; Fontoura, P.; Garren, H.; Hur, E.M.; Gupta, R.; Lee, L.Y.; Kidd, B.A.; Robinson, W.H.; et al. Treatment of autoimmune neuroinflammation with a synthetic tryptophan metabolite. Science 2005, 310, 850–855.
  77. Pemberton, L.A.; Kerr, S.J.; Smythe, G.; Brew, B.J. Quinolinic acid production by macrophages stimulated with IFN-gamma, TNF-alpha, and IFN-alpha. J. Interferon Cytokine Res. 1997, 17, 589–595.
  78. Sakurai, K.; Zou, J.P.; Tschetter, J.R.; Ward, J.M.; Shearer, G.M. Effect of indoleamine 2,3-dioxygenase on induction of experimental autoimmune encephalomyelitis. J. Neuroimmunol. 2002, 129, 186–196.
  79. Vécsei, L.; Szalardy, L.; Fülöp, F.; Toldi, J. Kynurenines in the CNS: Recent advances and new questions. Nat. Rev. Drug Discov. 2013, 12, 64–82.
  80. Tanaka, M.; Vécsei, L. Monitoring the Redox Status in Multiple Sclerosis. Biomedicines 2020, 8, 406.
  81. Kwidzinski, E.; Bechmann, I. IDO expression in the brain: A double-edged sword. J. Mol. Med. (Berl) 2007, 85, 1351–1359.
  82. Erabi, H.; Okada, G.; Shibasaki, C.; Setoyama, D.; Kang, D.; Takamura, M.; Yoshino, A.; Fuchikami, M.; Kurata, A.; Kato, T.A.; et al. Kynurenic acid is a potential overlapped biomarker between diagnosis and treatment response for depression from metabolome analysis. Sci. Rep. 2020, 10, 16822.
  83. Carrillo-Mora, P.; Pérez-De la Cruz, V.; Estrada-Cortés, B.; Toussaint-González, P.; Martínez-Cortéz, J.A.; Rodríguez-Barragán, M.; Quinzaños-Fresnedo, J.; Rangel-Caballero, F.; Gamboa-Coria, G.; Sánchez-Vázquez, I.; et al. Serum Kynurenines Correlate with Depressive Symptoms and Disability in Poststroke Patients: A Cross-sectional Study. Neurorehabilit. Neural Repair 2020.
  84. Ulivieri, M.; Wierońska, J.M.; Lionetto, L.; Martinello, K.; Cieslik, P.; Chocyk, A.; Curto, M.; Di Menna, L.; Iacovelli, L.; Traficante, A.; et al. The Trace Kynurenine, Cinnabarinic Acid, Displays Potent Antipsychotic-Like Activity in Mice and Its Levels Are Reduced in the Prefrontal Cortex of Individuals Affected by Schizophrenia. Schizophr. Bull. 2020.
  85. Małgorzata, P.; Paweł, K.; Iwona, M.L.; Brzostek, T.; Andrzej, P. Glutamatergic dysregulation in mood disorders: Opportunities for the discovery of novel drug targets. Expert Opin. Ther. Targets 2020, 3, 1–23.
  86. Koola, M.M. Alpha7 nicotinic-N-methyl-d-aspartate hypothesis in the treatment of schizophrenia and beyond. Hum. Psychopharmacol. 2020.
  87. Kim, E.Y.; Ahn, H.-S.; Lee, M.Y.; Yu, J.; Yeom, J.; Jeong, H.; Min, H.; Lee, H.J.; Kim, K.; Ahn, Y.M. An Exploratory Pilot Study with Plasma Protein Signatures Associated with Response of Patients with Depression to Antidepressant Treatment for 10 Weeks. Biomedicines 2020, 8, 455.
  88. Tanaka, M.; Telegdy, G. Neurotransmissions of antidepressant-like effects of neuromedin U-23 in mice. Behav. Brain Res. 2014, 259, 196–199.
  89. Tanaka, M.; Kádár, K.; Tóth, G.; Telegdy, G. Antidepressant-like effects of urocortin 3 fragments. Brain Res. Bull. 2011, 84, 414–418.
  90. Telegdy, G.; Tanaka, M.; Schally, A.V. Effects of the LHRH antagonist Cetrorelix on the brain function in mice. Neuropeptides 2009, 43, 229–234.
  91. Tanaka, M.; Schally, A.V.; Telegdy, G. Neurotransmission of the antidepressant-like effects of the growth hormone-releasing hormone antagonist MZ-4-71. Behav. Brain Res. 2012, 228, 388–391.
  92. Rog, J.; Błażewicz, A.; Juchnowicz, D.; Ludwiczuk, A.; Stelmach, E.; Kozioł, M.; Karakula, M.; Niziński, P.; Karakula-Juchnowicz, H. The Role of GPR120 Receptor in Essential Fatty Acids Metabolism in Schizophrenia. Biomedicines 2020, 8, 243.
  93. López-Gambero, A.J.; Sanjuan, C.; Serrano-Castro, P.J.; Suárez, J.; Rodríguez de Fonseca, F. The Biomedical Uses of Inositols: A Nutraceutical Approach to Metabolic Dysfunction in Aging and Neurodegenerative Diseases. Biomedicines 2020, 8, 295.
  94. Cantón-Habas, V.; Rich-Ruiz, M.; Romero-Saldaña, M.; Carrera-González, M.P. Depression as a Risk Factor for Dementia and Alzheimer’s Disease. Biomedicines 2020, 8, 45.
  95. Kowalska, K.; Krzywoszański, Ł.; Droś, J.; Pasińska, P.; Wilk, A.; Klimkowicz-Mrowiec, A. Early Depression Independently of Other Neuropsychiatric Conditions, Influences Disability and Mortality after Stroke (Research Study—Part of PROPOLIS Study). Biomedicines 2020, 8, 509.
  96. Vargas, A.S.; Luís, Â.; Barroso, M.; Gallardo, E.; Pereira, L. Psilocybin as a New Approach to Treat Depression and Anxiety in the Context of Life-Threatening Diseases—A Systematic Review and Meta-Analysis of Clinical Trials. Biomedicines 2020, 8, 331.
  97. Park, S.; Bak, A.; Kim, S.; Nam, Y.; Kim, H.; Yoo, D.-H.; Moon, M. Animal-Assisted and Pet-Robot Interventions for Ameliorating Behavioral and Psychological Symptoms of Dementia: A Systematic Review and Meta-Analysis. Biomedicines 2020, 8, 150.
More
This entry is offline, you can click here to edit this entry!
Video Production Service