The tryptophan catabolite or kynurenine pathway in Alzheimer’s disease: a systematic review and meta-analysis =============================================================================================================== * Abbas F. Almulla * Thitiporn Supasitthumrong * Arisara Amrapala * Chavit Tunvirachaisakul * Al-Karrar Kais Abdul Jaleel * Gregory Oxenkrug * Hussein K. Al-Hakeim * Michael Maes ## Abstract **Background** Alzheimer’s disease (AD), characterized by gradual brain dysfunction and memory loss, is one of the major elderly health issues worldwide. Neuroinflammation and increased oxidative stress contribute to the pathophysiology of AD, thereby presumably inducing tryptophan (TRP) degradation through the TRP catabolite (TRYCAT) pathway. **Objectives** To delineate the activity of the TRYCAT pathway along with levels of TRP and tryptophan catabolites (TRYCATs) in AD patients. **Methods** We employed PubMed, Google Scholar, Web of Science, and SciFinder to obtain the relevant articles through a search process lasting the entire February 2022. We found 19 eligible articles which involved 738 patients and 665 healthy controls. **Results** Our result revealed a significant difference (*p* = 0.008) in the kynurenine (KYN)/TRP ratio (standardized mean difference, SMD = 0.216, 95% confidence interval, CI: 0.057; 0.376), and a significant decrease in TRP in AD patients (SMD = -0.520, 95% CI: -0.738; -0.302, p < 0.0001). Moreover, we also found a significant increase in the central nervous system (CNS, brain and cerebrospinal fluid, CSF) kynurenic acid (KA)/KYN ratio but not in peripheral blood, as well as a significant decrease in plasma KA and xanthurenic acid (XA) in the CNS and blood. **Conclusions** AD is characterized by TRP depletion but not by an overactivity of the TRYCAT pathway. IDO-induced production of neurotoxic TRYCATs is not a key factor in the pathophysiology of AD. Keywords * Alzheimer’s disease * Kynurenine pathway * TRYCATs * tryptophan catabolites ## Introduction Alzheimer’s disease (AD) is one of the major elderly health issues worldwide, and is a progressive neurodegenerative disease with gradual memory deterioration and behavior aberrations (Reddy and Beal 2008, Cornutiu 2015, Morris, Berk et al. 2019). The pathophysiology of AD is associated with increased precipitation of amyloid-β (Aβ), aggregation of phosphorylated tau proteins, and apoptosis of neurons in the central nervous system (CNS) (Mattson 2004, LaFerla, Green et al. 2007, O’Brien and Wong 2011). However, these pathophysiological findings are possibly non-unique contributors to the AD pathophysiology because normal individuals without cognitive symptoms may also exhibit elevations in amyloid deposits, as indicated by positron emission tomography studies (Chételat, La Joie et al. 2013). Moreover, clinical severity in AD patients is not significantly correlated with amyloid deposition. Hence, the pathophysiological picture of AD remains ambiguous (Chételat, La Joie et al. 2013, Morris, Clark et al. 2014). Several postmortem studies have indicated the implication of an imbalanced immune system in the pathological state of AD. These studies reported that amyloid plaques in the brain tissues of AD patients showed an aggregate of interleukin (IL)-1β (Griffin, Stanley et al. 1989), IL-6 (Hüll, Berger et al. 1996, Bonaccorso, Lin et al. 1998), and tumor growth factor-β (TGF-β) (van der Wal, Gómez-Pinilla et al. 1993). Thus, AD is associated with neuroinflammation followed by upregulated microglia and astrocytes, and increased neuroinflammatory markers (Heneka, Carson et al. 2015). In addition, in cerebrospinal fluid (CSF) and plasma of AD patients, elevated levels of pro and anti-inflammatory cytokines are observed, namely IL-1β, IL-6, and tumor necrosis factor (TNF)-α, and IL-1 receptor antagonist (IL-1ra) and IL-10, respectively (Maes, DeVos et al. 1999, Brosseron, Krauthausen et al. 2014). High plasma levels of cortisol and interferon-gamma (IFN-γ) were found especially in the milder phases of AD patients (Zvěřová, Fišar et al. 2013, Belkhelfa, Rafa et al. 2014). Neuronal cells are strongly influenced by oxidative and nitrosative stress (O&NS), much more than peripheral tissues, due to the increased rate of metabolism and relative lower levels of antioxidants (Maes, Galecki et al. 2011). Moreover, amyloid plaque aggregations cause neurotoxicity because they drive microglia and astrocytes overactivity, releasing pro-inflammatory cytokines and reactive oxygen species (ROS), consequently causing O&NS in the brain (Moneim 2015). Administration of antioxidants may help prevent disease progression and decrease risk of developing AD (Moneim 2015). Furthermore, in AD patients, a large body of evidence has confirmed the presence of increased plasma levels of lipopolysaccharides (LPS) (Zhang, Miller et al. 2009, Zhan, Stamova et al. 2016, Zhao, Jaber et al. 2017, Kim, Kim et al. 2021), which intrinsically has toxic effects and promotes O&NS and immune activation (Halawa, A El-Adl et al. 2018). Immune activation with increased production of IFN-γ and M1 macrophage cytokines, and increased ROS/O&NS and LPS may induce indoleamine 2,3-dioxygenase, the key regulatory enzyme in the tryptophan catabolite (TRYCAT) pathway (Maes, Leonard et al. 2011). IDO activation may cause a reduction in tryptophan (TRP) levels and increased TRYCAT levels as shown in **Figure 1** (Maes, Meltzer et al. 1993, Smith and Maes 1995, Kanchanatawan, Sirivichayakul et al. 2018). Indeed, studies on AD and mild cognitive impairment (MCI) patients display a high kynurenine (KYN)/TRP ratio (Widner, Leblhuber et al. 2000, Greilberger, Fuchs et al. 2010). Thus, TRP will be diverted from the production of vital derivates, such as serotonin, which maintains neuroplasticity and protection against neuronal damage (Croonenberghs, Verkerk et al. 2005, Radulescu, Dragoi et al. 2021) to the TRYCAT pathway, potentially producing neurotoxic metabolites, including quinolinic acid (QA), xanthurenic acid (XA), 3-hydroxykynurenine (3HK), picolinic acid (PA), and 3-hydroxyanthranilic acid (3HA). ![Figure 1:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/03/21/2022.03.18.22272608/F1.medium.gif) [Figure 1:](http://medrxiv.org/content/early/2022/03/21/2022.03.18.22272608/F1) Figure 1: Summary of the TRYCAT pathway in AD IFN-γ: Interferon-Gamma, IL-6: Interleukin 6, IL-1β: Interleukin-1 beta, O&NS: Oxidative and nitrosative stress, LPS: Lipopolysaccharides, CNS: Central nervous system, IDO: Indoleamine 2,3 dioxygenase, TDO: Tryptophan 2,3-dioxygenase, KAT: Kynurenine Aminotransferase, KMO: Kynurenine 3-monooxygenase, KYNU: Kynureninase, TRP: Tryptophan, KYN: Kynurenine, KA: Kynurenic Acid, 3HK: 3-Hydroxykynurenine, AA: Anthranilic Acid, XA: Xanthurenic Acid, 3HA: 3-Hydroxyanthranilic Acid, PA: Picolinic Acid, QA: Quinolinic Acid. Some of these tryptophan catabolites (TRYCATs), namely 3HA, 3HK, and QA, are pro-oxidants and induce O&NS via generation of ROS and reactive nitrogen species (RNS) such as superoxide ion and peroxides, whilst also boosting copper-dependent alpha-crystallin cross-linking (Goldstein, Leopold et al. 2000, Smith, Smith et al. 2009, Zadori, Veres et al. 2018). Moreover, these metabolites mediate cell damage due to the formation of free radicals (Dykens, Sullivan et al. 1987, Okuda, Nishiyama et al. 1998). QA may also cause lipid peroxidation as a result of producing ROS (Rios and Santamaria 1991, Santamaría, Galván-Arzate et al. 2001) and has been reported to exhibit excitatory properties on N-methyl-D-aspartate (NMDA) receptors (Kanchanatawan, Sirivichayakul et al. 2018) and pro-inflammatory effects, particularly in the microglia thereby augmenting the inflammatory, oxidative stress and neurotoxicity effects (Maes, Leonard et al. 2011). On the other hand, various TRYCATs also have neuroprotective roles, such as kynurenic acid (KA), which regulates NMDA receptor functions (Morris, Carvalho et al. 2016), whilst anthranilic acid (AA) impedes the production of QA and PA (Guillemin, Cullen et al. 2007, Almulla and Maes 2022). Furthermore, XA may have neuroprotective effects via reducing vesicular glutamate transport (VGLUT), downregulating NMDA receptors, and attenuating postsynaptic excitatory potency (Kanchanatawan, Sirivichayakul et al. 2018). Nonetheless, in AD there is no systematic review and meta-analysis examining TRYCAT pathway activity. Hence, in the current study, we perform a comprehensive meta-analysis and systematic review of TRP and TRYCAT data in patients with AD compared to healthy controls. ## Material and Methods We employed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) 2020 criteria, the Cochrane Handbook for Systematic Reviews and Interventions guidelines, and Meta-Analyses of Observational Studies in Epidemiology (MOOSE) guidelines to conduct the present meta-analysis. We investigate TRP along with TRYCATs levels, whilst IDO, KAT, and KMO enzyme activity were examined through their indices, namely, KYN/TRP and (KA+KYN)/TRP, KA/KYN and KA/(KYN+TRP), and 3HK/KYN ratios, respectively. ### Search strategy We began the database search process on January 1st, 2022. Specific keywords and AD-associated MESH terms were entered into the electronic databases, namely PubMed/MEDLINE, Google Scholar, Web of Science, and SciFinder. These keywords, along with the terms, covered TRP, TRYCATs, and AD, all of which are displayed in the electronic supplementary file (ESF), Table 1. The reference lists of prior reviews and grey literature were also examined to ensure that all relevant papers were included. By the end of the month, all relevant articles were collected. View this table: [Table 1.](http://medrxiv.org/content/early/2022/03/21/2022.03.18.22272608/T1) Table 1. The outcomes profile and number of AD patients and healthy control along with the side of standardized mean difference (SMD) and the 95% confidence intervals with respect to zero SMD ### Eligibility Criteria Peer-reviewed journals and English language were the primary criteria for selecting eligible articles for the study. We did, nevertheless, assess manuscripts in other languages, including Thai, French, Spanish, German, Italian, and Arabic. All included articles were compliant with the following inclusion criteria: a) observational case-control studies that employed serum, plasma, CSF, and brain tissues to investigate the levels of TRP and TRYCATs in patients with AD compared to healthy controls; b) studies that recruited cognitively normal individuals as a control group and who were free of any active psychiatric illness; and c) articles that presented numerical values of mean with standard deviation (SD) or mean standard error (SEM) for TRP and TRYCATs. Based on the specified criteria, the following studies were excluded a) systematic and narrative review articles, as well as meta-analyses; b) animal and genetic studies, and duplicate articles; c) studies which used saliva and hair to assay TRYCATs; d) studies reporting other types of dementia or pre-clinical AD patients; and e) articles that did not reveal mean SD/SE of the assessed TRP and TRYCATs. We emailed the authors to request the mean and SD of the biomarkers if they published geometric means, median (interquartile range, range), or showed data as a graph. In the absence of a response from the authors, we used Wan and Wang’s estimation method to compute mean SD from the median and interquartile range or range. Furthermore, Web Plot Digitizer was used to extract quantitative data from graphs showing means and SD/SEM of TRYCATs ([https://automeris.io/WebPlotDigitizer/](https://automeris.io/WebPlotDigitizer/)). ### Primary and secondary outcomes The KYN/TRP and (KYN+KA)/TRP ratios were utilized to examine the primary outcome, which was IDO enzyme activity in AD patients versus healthy controls. Secondary outcomes included the KAT enzyme indices, namely KA/KYN and KA/(KYN+TRP) ratios, the KMO enzyme proxy 3HK/KYN ratio, and the neurotoxic index (KYN+3HK+QA+XA+PA)/(KA+AA). The levels of TRP, KYN, KA, 3HK, AA, XA, and QA were also assessed in patients with AD versus healthy controls. ### Screening and data extraction The first author (AA) carried out the preliminary review to determine eligible studies for inclusion by inspecting the title and abstract of the searched articles. The full texts of the eligible studies were then downloaded after screening with the pre-specified exclusion criteria. After the first author finished inputting the data in an Excel spreadsheet, the second author (TS) reviewed the accuracy of the data. The last author (MM) was consulted for any equivocal findings. A pre-specified excel spreadsheet was used to extract and organize the required data in this study. This data file comprised the authors’ names, publication dates, numerical data of TRP and TRYCATs, the number of recruited individuals either as AD patients or healthy controls, background variables such as age and number of males and females in each group, statistical data such as mean (SD/SE), sample type, mini-mental state examination (MMSE) score, the latitude of the country where the study was conducted, and the quality and redpoint scores of the included studies. The methodological quality of included studies was evaluated using the prior known “immune confounder scales (ICS)” after certain modifications were made by the last author (MM), as shown in ESF, Table 3. We assessed the essential quality determinants by the ICS, namely sample size, controlling of the confounders, and the time at which the sample was taken. The range of the overall quality score was 0 to 10, with 10 being the best quality. An uncontrolled study design and insufficient adjustment of major confounders were addressed in the redpoint score scale, which has a range of 0 to 26, with higher scores implicating a lack of control and higher exposure of the TRYCATs results to many types of bias, including biological and analytical. ### Data analysis We performed the present meta-analysis using the CMA V3 software and complied with all PRISMA criteria. As a minimum, three studies reporting each TRYCAT was required to carry out a meta-analysis. **Table 1** shows the outcome profile of the TRYCATs, which were addressed in the current systematic review and meta-analysis. When several biomarkers representing the same profile were reported in the same research, a synthetic score was calculated using the mean of the results, assuming dependency. The computed scores, which reflect different profiles, as well as solitary TRYCATs were compared between AD patients and healthy controls. We examined the activity of IDO, KAT and KMO enzymes through their proxies as follows: a) IDO was assessed using KYN/TRP and (KYN+KA)/TRP ratios by setting the effect size direction of KYN (KA) as positive for increased KYN (KA) and positive for lowered TRP levels in AD; and b) KAT activity was determined by the KA/KYN and KA/(KYN+TRP) ratios by specifying a positive effect size direction for increased KA in AD patients and a negative direction for TRP and KYN in the meta-analysis. The random-effects model with restricted maximum-likelihood was utilized in this investigation on the premise that population characteristics varied between studies. The effect size was calculated using the standardized mean difference (SMD) with 95% confidence intervals (95% CI), and two-tailed, *p* < 0.05 was considered statistically significant. SMD values of 0.8, 0.5 and 0.2 indicate large, moderate, and modest effect sizes, respectively (Cohen 2013). We computed tau-squared, as previously indicated, to evaluate the heterogeneity in addition to the Q and I2 values (Vasupanrajit, Jirakran et al. 2021, Vasupanrajit, Jirakran et al. 2021). To test the strength of the findings, we performed a sensitivity analysis by using the “removing-one-study approach.” Subgroup analysis was employed in this meta-analysis to examine the differences between brain tissues, CSF, serum, and plasma (Almulla, Vasupanrajit et al. 2021), and each of these groups were considered as the unit of analysis. In the case that there were no significant differences between brain tissues and CSF, they were combined to a single entity called the central nervous system (CNS). The fail-safe N method, Kendall tau with continuity correction, and Egger’s regression intercept (both using a one-tailed *p*-value) were used to reveal the influence of small study effects. We accounted for the modified effect size computed by imputing the missing studies by the Duval and Tweedie’s trim-and-fill method in case of significant asymmetry reported by Egger’s test. We conducted a meta-regression in the current study to assess the sources of heterogeneity. ## Results ### Search findings PRISMA flowchart **Figure 2** shows the detailed results of the search processes in the different databases and the standards used for inclusion-exclusion of research articles. Based on prespecified terms and keywords listed in ESF, Table 1, we initially identified 24 full-text papers. Five articles were removed due to reasons mentioned in the ESF, Table 5. Accordingly, 19 full-text articles were considered in the current meta-analysis (Tohgi, Abe et al. 1992, Tohgi, Abe et al. 1995, Bonaccorso, Lin et al. 1998, Baran, Jellinger et al. 1999, Widner, Leblhuber et al. 2000, Hartai, Juhasz et al. 2007, Greilberger, Fuchs et al. 2010, Gulaj, Pawlak et al. 2010, Schwarz, Guillemin et al. 2013, Wennstrom, Nielsen et al. 2014, Rommer, Fuchs et al. 2016, Giil, Midttun et al. 2017, Oxenkrug, van der Hart et al. 2017, Jacobs, Lim et al. 2019, Sorgdrager, Vermeiren et al. 2019, van der Velpen, Teav et al. 2019, Gonzalez-Sanchez, Jimenez et al. 2020, Whiley, Chappell et al. 2021, Willette, Pappas et al. 2021). ![Figure 2:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/03/21/2022.03.18.22272608/F2.medium.gif) [Figure 2:](http://medrxiv.org/content/early/2022/03/21/2022.03.18.22272608/F2) Figure 2: The PRISMA flow chart We included case-control studies in this meta-analysis, resulting in a total investigation of 1,403 individuals, distributed as 738 AD patients and 665 healthy controls. We examined the subjects’ TRP and TRYCATs levels in 8 studies within the CNS (7 CSF and 1 brain tissue), 8 in plasma, and 7 in serum. Four studies measured central and peripheral levels of the TRP and TRYCATs. In the serum, TRP and TRYCAT levels were examined in 582 participants (321 AD patients versus 261 healthy controls), 487 participants (244 AD patients versus 243 healthy controls) were evaluated in plasma studies, and 335 participants (173 AD patients versus 162 healthy controls) were investigated in CNS studies. No significant differences were found between central and peripheral outcomes except in KA levels and KA/KYN ratio. Additionally, there were no significant differences between serum and plasma. Therefore, we combined all serum/plasma studies and pooled the overall effect size. High-performance liquid chromatography (HPLC) was the most used analytic technique (reported in 10 studies), followed by LC (2 studies), while the remaining studies employed different techniques, as listed in ESF, Table 5. Austria contributed with 4 studies, while Belgium, Japan, Sweden, and the USA participated with 2 studies, and 1 study was reported by research groups from Germany, Hungary, Norway, Poland, Switzerland, UK, and Spain. The redpoint and quality scores were 11 (median) (min=10, max=12.5) and 5.5 (min=2.5 max=9), respectively. The subjects’ ages ranged from 63 to 81 years. ### Primary outcome variables #### KYN/TRP and (KYN+KA)/TRP ratios In the current meta-analysis, we included 22 studies concerning the KYN/TRP ratio: 5 studies with 95% CI that were entirely on the positive side of zero, whereas no 95% CI were entirely on the negative side of zero. The 95% CI of the other 17 studies intersected with zero, with 10 studies showing a positive SMD value, and 7 studies showing SMD values less than zero as displayed in **Table 1**. The overall results in **Table 2** indicate a significant difference with modest effect size in the KYN/TRP ratio between AD patients and healthy controls, as illustrated in the forest plot (**Figure 3)**. Publication bias was observed in 2 studies to the left, and the adjusted estimate point changed to 0.165 (95%CI:-0.001;0.332) after imputing those 2 studies. View this table: [Table 2.](http://medrxiv.org/content/early/2022/03/21/2022.03.18.22272608/T2) Table 2. Results of meta-analysis performed on several outcomes (TRYCATs) variables with combined different media and separately. ![Figure 3:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/03/21/2022.03.18.22272608/F3.medium.gif) [Figure 3:](http://medrxiv.org/content/early/2022/03/21/2022.03.18.22272608/F3) Figure 3: Forest plot of the kynurenine/tryptophan (KYN/TRP) ratio reflecting indoleamine-2,3-dioxygenase (IDO) activity with results of a meta-analysis conducted on 22 studies. The forest plot for the (KYN+KA)/TRP ratio is shown in ESF, Figure 1. **Table 2** indicates a significant increase with a very modest effect size in AD compared to healthy controls. **Table 3** indicates a possible bias with 8 studies to the left side. Imputing these studies alters the SMD to 0.015 (95%CI: -0.177;0.147), which is no longer significant. View this table: [Table 3.](http://medrxiv.org/content/early/2022/03/21/2022.03.18.22272608/T3) Table 3. Results on publication bias. ### Secondary outcome variables #### The KA/KYN and KA/(KYN+TRP) ratios We found a high heterogeneity in the KA/KYN results and, therefore, performed subgroup analyses that revealed a significant difference (*p*=0.034) between CNS and peripheral levels. **Table 2** and ESF, Figure 2 shows that the KA/KYN ratio in the CNS of AD patients significantly increased compared to healthy controls with a moderate effect size (SMD=0.466). Contrastingly, the results in serum and plasma were not significant. **Table 3** indicates a possible bias in the CNS results with 1 study to the right side missing and after imputation, the SMD increased to 0.615 (95%CI: 0.112; 1.118). The results in plasma and serum showed a possible bias with 1 study missing on the left side for plasma, and 3 studies on the right side for serum. After adjusting the effect sizes by imputing for those studies, the results remained significant. The forest plot of the KA/(KYN+TRP) ratio (ESF, Figure 3) and **Table 2** show a significant increase with a small effect size (SMD= 0.217) in AD patients compared to controls. Kendall’s tau and Egger’s regression (**Table 3**) indicate possible bias with 4 studies missing on the right side. After imputing these data, the difference was no longer significant (SMD= 0.147; 95%CI: -0.001; 0.294). #### The 3HK/KYN ratio ESF, Figure 4, and **Table 2** show that there is no significant difference in the 3HK/KYN between AD patients and healthy controls. **Table 3** revealed a possible bias with 3 studies missing on the right side. ![Figure 4:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/03/21/2022.03.18.22272608/F4.medium.gif) [Figure 4:](http://medrxiv.org/content/early/2022/03/21/2022.03.18.22272608/F4) Figure 4: Forest plot with results of the meta-analysis performed on 20 studies reporting on tryptophan (TRP) in Alzheimer disease. #### The(KYN+3HK+XA+QA+PA)/(KA+AA) ratio There was no significant difference between AD patients and healthy controls in the neurotoxicity ratio (see **Table 2** and ESF, Figure 5). Publication bias was found with 4 studies missing on the left side. ### TRP and solitary TRYCATs **Table 1** shows the distributions of the 95% CI. **Table 2** shows that TRP was significantly decreased with a moderate effect size in patients with AD compared to healthy controls. Kendall’s tau and Egger’s regression indicate a significant bias with 2 studies missing on the right. The adjusted estimate point was still significant (**Table 3**). **Table 2** shows that there was no significant difference in KYN between AD patients and healthy controls (see **Table 2** and ESF, Figure 6). AD patients showed a significant decrease in KA levels with a small effect size, however, there was a significant difference (*p*=0.043) between the central and peripheral results. In plasma, a significant decrease in KA was detected, whereas in CNS and serum no significant alterations were found. ESF, Figure 7 shows the forest plot of KA results. The results of 3HK studies (**Table 1** and **Table 2**) show no significant differences in 3HK between AD and controls. There were no significant differences in QA and AA between patients and controls, although there was a trend towards lowered AA levels in AD. XA results were pooled from 5 studies. **Table 2** and ESF, Figure 9 show a significant difference in XA between AD patients and healthy controls. After imputing 2 missing studies on the right, the SMD remained significant. ### Meta-regression analysis Meta-regression results are presented in ESF, Table 6. Briefly, it shows that gender, whether male/female or both, significantly affects most of the outcome data, namely KYN/TRP, (KA+KYN)/TRP, KA/KYN, (KYN+3HK+XA+QA+PA)/(KA+AA), TRP, and KYN. Age was found to influence the results of KYN/TRP, 3HK/KYN, 3HK, and TRP. The sample size affected the outcome of KYN/TRP, (KA+KYN)/TRP, and TRP. The other confounders, such as latitude, MMSE scores. etc., exerted some effect on single or multiple outcomes. ## Discussion The first major finding of this systematic review and meta-analysis is that both ratios reflecting the KYN/TRP ratio were significantly higher in AD than controls and, after adjusting for possible bias, all significance disappeared. Moreover, our meta-analysis did not show any changes in KYN in AD, whilst TRP levels were significantly decreased in AD patients. These results indicate normal IDO activity in AD patients and lowered availability of TRP, which may be explained by AD-associated reductions in levels of albumin that bind TRP (Maes, DeVos et al. 1999, Kim, Byun et al. 2020; Maes, Wauters et al. 1996). Furthermore, TRP availability to the brain is an age-sensitive process (Bonaccorso, Lin et al. 1998), which may contribute to low CNS levels of TRP in AD. In theory, reduced levels of TRP in AD may be due to malnutrition associated with cognitive dysfunction of AD patients (Giil, Midttun et al. 2017). Lowered levels of TRP in AD could be associated with increased behavioral and psychological symptoms of dementia (BPSD) including depression (Hood, Bell et al. 2005, Ruhé, Mason et al. 2007). Furthermore, TRP has neuroprotective effects and thus decreases may further aggravate neurodegenerative processes (Del Angel-Meza, Dávalos-Marín et al. 2011). The second major finding is that we detected a significant increase in the KA/KYN ratio in the CNS of AD patients but not in peripheral blood. In contrast, the KA/(KYN+TRP) ratio was significantly increased with a very small effect size in the total study group. Thus, these results suggest a mild activation of KAT enzyme in the CNS of patients with AD. Previous papers showed no changes in peripheral KAT activity (Hartai, Juhasz et al. 2007), although centrally KAT was activated (Baran, Jellinger et al. 1999). However, our findings revealed a significant decrease in plasma KA levels in AD without changes in CNS and serum levels. In line with our results, Hartai et al reported lowered levels of KA in CSF and plasma (Hartai, Juhasz et al. 2007). In addition, no change in CSF KA levels was found in AD (Wennstrom, Nielsen et al. 2014). Therefore, it is likely that the lowered KA production is the consequence of lowered levels of KYN and KAT activity as well. Furthermore, KA formation is influenced by aberrations in energy metabolism (Hodgkins and Schwarcz 1998), an early hallmark of AD (Hartai, Juhasz et al. 2007). KMO enzyme activity, as reflected by the 3HK/KYN ratio, was not significantly changed in AD. Apart from this, our results also indicate no significant change in overall 3HK levels, confirming our KMO results. Previous studies reported no changes in 3HK levels in AD patients compared to controls, in either central or peripheral levels (Baran, Jellinger et al. 1999, Gulaj, Pawlak et al. 2010, Jacobs, Lim et al. 2019, Sorgdrager, Vermeiren et al. 2019). KMO enzyme requires NADPH as a coenzyme (Hughes, Guner et al. 2022) and, therefore, the overactivity of NADPH oxidase enzyme consumes NADPH in AD patients (Fragoso-Morales, Correa-Basurto et al. 2021), which may cause attenuated KMO enzyme activity. The fourth major finding of the present study is that there was no significant difference in the TRYCAT neurotoxicity (KYN+3HK+XA+QA+PA) / (KA+AA) ratio between AD patients and controls. Furthermore, our results showed a significant decrease in XA levels, one of the neurotoxic TRYCATs, in AD versus controls. Previous studies reported low XA levels in AD patients (Giil, Midttun et al. 2017, Sorgdrager, Vermeiren et al. 2019). The decreased XA in AD may in part be ascribed to lowered levels of 3HK and lowered TRYCAT pathway activity because of lowered TRP levels. In addition, XA may decrease in response to high levels of free radicals which are detected in AD (Giil, Midttun et al. 2017). We also found that QA was not significantly altered in AD patients compared to controls. For example, Greilberger et al and Schwarz reported no changes in QA levels (Greilberger, Fuchs et al. 2010, Schwarz, Guillemin et al. 2013). It is hypothesized that in AD, QA is potentially implicated in increased tau protein phosphorylation and contributes to the neurofibrillary tangles (Rahman, Ting et al. 2009, Lugo-Huitrón, Ugalde Muñiz et al. 2013). However, QA is largely produced from 3HK by activated microglia in the CNS and no changes in 3HK could be detected in our meta-analysis. Sorgdrager et al suggested that serum and CSF levels of QA in neurodegenerative disease may accumulate in response to aging (Sorgdrager, Vermeiren et al. 2019). Regardless, our results indicate that neurotoxicity due to increased levels of the neurotoxic TRYCATs does not play a role in AD. Finally, AA levels were significantly decreased in AD patients compared with controls, as previously reported in an early study (Oxenkrug, van der Hart et al. 2017). AA formation is regulated by activity of kynureninase, which catalyzes AA formation from KYN, an immediate substrate for biosynthesis of AA, 3HK and KYNA. Notably, kynureninase affinity to KYN is much lower than to KMO and KAT, and under physiological conditions KYN is mainly converted into 3HK and KYNA rather than into AA (Moroni 1999). Up-regulation of KMO, which catalyzes KYN formation into 3HK, might decrease the availability of KYN as a substrate for kynureninase and, consequently, down-regulate AA production. Elevation of 3HK serum levels might provide a mechanistical explanation of down-regulated AA formation (Oxenkrug, van der Hart et al. 2017). However, the present meta-analysis reveals no significant differences in KYN and 3HK between AD and controls that might explain decreased plasma/serum AA levels. On the other hand, elevated plasma AA levels was the most powerful predictor of future development of incident dementia (Chouraki, Preis et al. 2017). Higher AA (and KYN) plasma levels were observed in high neocortical amyloid-beta load (NAL) than in noo-NAL female (but not male) patients prior to development of cognitive impairment, and AA (and KYN) levels individually and jointly predicted development of NAL(Chatterjee, Goozee et al. 2018). Notably, the alternative to IDO enzyme that catalyzes TRP conversion into KYN is tryptophan 2,3-dioxygenase 2 (TDO), which is activated by cortisol. Correlations between post-dexamethasone cortisol levels were observed in female, but not male, AD patients (Oxenkrug, Gurevich et al. 1989). ### Limitations Limitations regarding the current systematic review and meta-analysis are as follows: First, most included studies did not delineate the exact stage of AD, and we were, therefore, not able to examine the stage-related changes in TRYCATs levels. Second, there was a lack of information on the patients’ treatments which might affect TRYCATs levels. Third, the lack of postmortem studies and small sample sizes concerning CSF studies restrict our ability to draw conclusions on the TRYCAT status in the brain. Lastly, most studies did not report cognitive impairments and psychiatric comorbidities, which may further change TRYCAT pathway activity (Almulla, Vasupanrajit et al. 2021). ## Conclusions Figure 1. summarizes the results of the present study. Our findings indicate that the availability of TRP is reduced in patients with AD, although there are no signs that IDO is activated. Lowered TRP availability to the brain may be explained by lowered plasma albumin in AD. Moreover, there is a trend towards lowered AA, KA, and XA in AD, which may in part be explained by lowered levels of TRP. Since the composite TRYCAT neurotoxicity index was not increased in AD, we conclude that TRYCATs may not contribute to the neurotoxic pathophysiology of AD. ## Supporting information supplementary file [[supplements/272608_file02.pdf]](pending:yes) ## Data Availability On a reasonable request and after the authors have fully used the dataset, the employed dataset (excel file) developed for this meta-analysis will be accessible from MM. ## Declaration of Competing Interests The authors declare that this work is free from any conflict of interest. ## Ethical approval and consent to participate Not applicable. ## Consent for publication Not applicable. ## Availability of data and materials On a reasonable request and after the authors have fully used the dataset, the employed dataset (excel file) developed for this meta-analysis will be accessible from MM. ## Funding The study was funded by the C2F program, Chulalongkorn University, Thailand. ## Author’s contributions All the authors contributed to the writing of this work. AA and MM designed the study. AA and TS collected the data. AA and MM performed the statistical analysis. ## Acknowledgments Not applicable. ## Footnotes * E-mail addresses: Abbass.chem.almulla1991{at}gmail.com * Thitiporn.S{at}chula.ac.th * lily.amrapala{at}gmail.com * Chavit.tun{at}gmail.com * Alkkais50{at}gmail.com * goxenkrug{at}tuftsmedicalcenter.org * headm2010{at}yahoo.com * Received March 18, 2022. * Revision received March 18, 2022. * Accepted March 21, 2022. * © 2022, Posted by Cold Spring Harbor Laboratory This pre-print is available under a Creative Commons License (Attribution-NonCommercial-NoDerivs 4.0 International), CC BY-NC-ND 4.0, as described at [http://creativecommons.org/licenses/by-nc-nd/4.0/](http://creativecommons.org/licenses/by-nc-nd/4.0/) ## References 1. Almulla, A. F. and M. Maes (2022). “The tryptophan catabolite or kynurenine pathway’s role in major depression.” Preprints 2022010134. 2. Almulla, A. F., A. Vasupanrajit, C. Tunvirachaisakul, H. K. Al-Hakeim, M. Solmi, R. Verkerk and M. Maes (2021). “The tryptophan catabolite or kynurenine pathway in schizophrenia: meta-analysis reveals dissociations between central, serum and plasma compartments.” medRxiv: 2021.2012.2016.21267905. 3. Baran, H., K. Jellinger and L. Deecke (1999). “Kynurenine metabolism in Alzheimer’s disease.” J Neural Transm (Vienna) 106(2): 165–181. 4. Belkhelfa, M., H. Rafa, O. Medjeber, A. Arroul-Lammali, N. Behairi, M. Abada-Bendib, M. Makrelouf, S. Belarbi, A. N. Masmoudi, M. Tazir and C. Touil-Boukoffa (2014). “IFN-γ and TNF-α are involved during Alzheimer disease progression and correlate with nitric oxide production: a study in Algerian patients.” J Interferon Cytokine Res 34(11): 839–847. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1089/jir.2013.0085&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24831467&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) 5. Bonaccorso, S., A. Lin, C. Song, R. Verkerk, G. Kenis, E. Bosmans, S. Scharpe, M. Vandewoude, A. Dossche and M. Maes (1998). “Serotonin-immune interactions in elderly volunteers and in patients with Alzheimer’s disease (DAT): lower plasma tryptophan availability to the brain in the elderly and increased serum interleukin-6 in DAT.” Aging (Milano) 10(4): 316–323. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=9825023&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) 6. Brosseron, F., M. Krauthausen, M. Kummer and M. T. Heneka (2014). “Body fluid cytokine levels in mild cognitive impairment and Alzheimer’s disease: a comparative overview.” Mol Neurobiol 50(2): 534–544. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s12035-014-8657-1&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24567119&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) 7. Chatterjee, P., K. Goozee, C. K. Lim, I. James, K. Shen, K. R. Jacobs, H. R. Sohrabi, T. Shah, P. R. Asih, P. Dave, C. ManYan, K. Taddei, D. B. Lovejoy, R. Chung, G. J. Guillemin and R. N. Martins (2018). “Alterations in serum kynurenine pathway metabolites in individuals with high neocortical amyloid-beta load: A pilot study.” Sci Rep 8(1): 8008. 8. Chételat, G., R. La Joie, N. Villain, A. Perrotin, V. de La Sayette, F. Eustache and R. Vandenberghe (2013). “Amyloid imaging in cognitively normal individuals, at-risk populations and preclinical Alzheimer’s disease.” NeuroImage: Clinical 2: 356–365. 9. Chouraki, V., S. R. Preis, Q. Yang, A. Beiser, S. Li, M. G. Larson, G. Weinstein, T. J. Wang, R. E. Gerszten, R. S. Vasan and S. Seshadri (2017). “Association of amine biomarkers with incident dementia and Alzheimer’s disease in the Framingham Study.” Alzheimers Dement 13(12): 1327–1336. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jalz.2017.04.009&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28602601&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) 10. Cohen, J. (2013). Statistical power analysis for the behavioral sciences, Academic press. 11. Cornutiu, G. (2015). “The Epidemiological Scale of Alzheimer’s Disease.” J Clin Med Res7(9): 657–666. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.14740/jocmr2106w&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26251678&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) 12. Croonenberghs, J., R. Verkerk, S. Scharpe, D. Deboutte and M. Maes (2005). “Serotonergic disturbances in autistic disorder: L-5-hydroxytryptophan administration to autistic youngsters increases the blood concentrations of serotonin in patients but not in controls.” Life Sci 76(19): 2171–2183. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15733932&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) 13. Del Angel-Meza, A. R., A. J. Dávalos-Marín, L. L. Ontiveros-Martinez, G. G. Ortiz, C. Beas-Zarate, V. Chaparro-Huerta, B. M. Torres-Mendoza and O. K. Bitzer-Quintero (2011). “Protective effects of tryptophan on neuro-inflammation in rats after administering lipopolysaccharide.” Biomed Pharmacother 65(3): 215–219. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.biopha.2011.02.008&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21616633&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) 14. Dykens, J. A., S. G. Sullivan and A. Stern (1987). “Oxidative reactivity of the tryptophan metabolites 3-hydroxyanthranilate, cinnabarinate, quinolinate and picolinate.” Biochem Pharmacol 36(2): 211–217. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/0006-2952(87)90691-5&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=2949752&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1987F853800004&link_type=ISI) 15. Fragoso-Morales, L. G., J. Correa-Basurto and M. C. Rosales-Hernandez (2021). “Implication of Nicotinamide Adenine Dinucleotide Phosphate (NADPH) Oxidase and Its Inhibitors in Alzheimer’s Disease Murine Models.” Antioxidants (Basel) 10(2). 16. Giil, L. M., O. Midttun, H. Refsum, A. Ulvik, R. Advani, A. D. Smith and P. M. Ueland (2017). “Kynurenine Pathway Metabolites in Alzheimer’s Disease.” J Alzheimers Dis 60(2): 495–504. 17. Goldstein, L. E., M. C. Leopold, X. Huang, C. S. Atwood, A. J. Saunders, M. Hartshorn, J. T. Lim, K. Y. Faget, J. A. Muffat, R. C. Scarpa, L. T. Chylack, Jr.., E. F. Bowden, R. E. Tanzi and A. I. Bush (2000). “3-Hydroxykynurenine and 3-hydroxyanthranilic acid generate hydrogen peroxide and promote alpha-crystallin cross-linking by metal ion reduction.” Biochemistry 39(24): 7266–7275. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1021/bi992997s&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=10852726&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000087778300027&link_type=ISI) 18. Gonzalez-Sanchez, M., J. Jimenez, A. Narvaez, D. Antequera, S. Llamas-Velasco, A. H. Martin, J. A. M. Arjona, A. L. Munain, A. L. Bisa, M. P. Marco, M. Rodriguez-Nunez, D. A. Perez-Martinez, A. Villarejo-Galende, F. Bartolome, E. Dominguez and E. Carro (2020). “Kynurenic Acid Levels are Increased in the CSF of Alzheimer’s Disease Patients.” Biomolecules 10(4). 19. Greilberger, J., D. Fuchs, F. Leblhuber, M. Greilberger, R. Wintersteiger and E. Tafeit (2010). “Carbonyl proteins as a clinical marker in Alzheimer’s disease and its relation to tryptophan degradation and immune activation.” Clin Lab 56(9-10): 441–448. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21086789&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) 20. Griffin, W. S., L. C. Stanley, C. Ling, L. White, V. MacLeod, L. J. Perrot, C. L. White, 3rd. and C. Araoz (1989). “Brain interleukin 1 and S-100 immunoreactivity are elevated in Down syndrome and Alzheimer disease.” Proc Natl Acad Sci U S A 86(19): 7611–7615. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMDoiODYvMTkvNzYxMSI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIyLzAzLzIxLzIwMjIuMDMuMTguMjIyNzI2MDguYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 21. Guillemin, G. J., K. M. Cullen, C. K. Lim, G. A. Smythe, B. Garner, V. Kapoor, O. Takikawa and B. J. Brew (2007). “Characterization of the kynurenine pathway in human neurons.” J Neurosci 27(47): 12884–12892. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Njoiam5ldXJvIjtzOjU6InJlc2lkIjtzOjExOiIyNy80Ny8xMjg4NCI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIyLzAzLzIxLzIwMjIuMDMuMTguMjIyNzI2MDguYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 22. Gulaj, E., K. Pawlak, B. Bien and D. Pawlak (2010). “Kynurenine and its metabolites in Alzheimer’s disease patients.” Adv Med Sci 55(2): 204–211. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.2478/v10039-010-0023-6&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20639188&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) 23. Halawa, A. A., M. A El-Adl, M. F. Hamed, A. Z. Balboula, M. A. Elmetwally and B. DiK (2018). “Lipopolysaccharide Prompts Oxidative Stress and Apoptosis in Rats’ Testicular Tissue.” Journal of Veterinary Healthcare 1(3): 20–31. 24. Hartai, Z., A. Juhasz, A. Rimanoczy, T. Janaky, T. Donko, L. Dux, B. Penke, G. K. Toth, Z. Janka and J. Kalman (2007). “Decreased serum and red blood cell kynurenic acid levels in Alzheimer’s disease.” Neurochem Int 50(2): 308–313. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.neuint.2006.08.012&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17023091&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) 25. Heneka, M. T., M. J. Carson, J. E. Khoury, G. E. Landreth, F. Brosseron, D. L. Feinstein, A. H. Jacobs, T. Wyss-Coray, J. Vitorica, R. M. Ransohoff, K. Herrup, S. A. Frautschy, B. Finsen, G. C. Brown, A. Verkhratsky, K. Yamanaka, J. Koistinaho, E. Latz, A. Halle, G. C. Petzold, T. Town, D. Morgan, M. L. Shinohara, V. H. Perry, C. Holmes, N. G. Bazan, D. J. Brooks, S. Hunot, B. Joseph, N. Deigendesch, O. Garaschuk, E. Boddeke, C. A. Dinarello, J. C. Breitner, G. M. Cole, D. T. Golenbock and M. P. Kummer (2015). “Neuroinflammation in Alzheimer’s disease.” The Lancet Neurology 14(4): 388–405. 26. Hodgkins, P. S. and R. Schwarcz (1998). “Interference with cellular energy metabolism reduces kynurenic acid formation in rat brain slices: reversal by lactate and pyruvate.” EurJ Neurosci 10(6): 1986–1994. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1046/j.1460-9568.1998.00208.x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=9753086&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) 27. Hood, S. D., C. J. Bell and D. J. Nutt (2005). “Acute tryptophan depletion. Part I: rationale and methodology.” Aust N Z J Psychiatry 39(7): 558–564. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1080/j.1440-1614.2005.01627.x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15996136&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000230057200005&link_type=ISI) 28. Hughes, T. D., O. F. Guner, E. C. Iradukunda, R. S. Phillips and J. P. Bowen (2022). “The Kynurenine Pathway and Kynurenine 3-Monooxygenase Inhibitors.” Molecules 27(1). 29. Hüll, M., M. Berger, B. Volk and J. Bauer (1996). “Occurrence of interleukin-6 in cortical plaques of Alzheimer’s disease patients may precede transformation of diffuse into neuritic plaques.” Ann N Y Acad Sci 777: 205–212. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=8624085&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1996BF30Z00026&link_type=ISI) 30. Jacobs, K. R., C. K. Lim, K. Blennow, H. Zetterberg, P. Chatterjee, R. N. Martins, B. J. Brew, G. J. Guillemin and D. B. Lovejoy (2019). “Correlation between plasma and CSF concentrations of kynurenine pathway metabolites in Alzheimer’s disease and relationship to amyloid-beta and tau.” Neurobiol Aging 80: 11–20. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) 31. Kanchanatawan, B., S. Sirivichayakul, K. Ruxrungtham, A. F. Carvalho, M. Geffard, G. Anderson and M. Maes (2018). “Deficit Schizophrenia Is Characterized by Defects in IgM-Mediated Responses to Tryptophan Catabolites (TRYCATs): a Paradigm Shift Towards Defects in Natural Self-Regulatory Immune Responses Coupled with Mucosa-Derived TRYCAT Pathway Activation.” Mol Neurobiol 55(3): 2214–2226. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s12035-017-0465-y&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28290151&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) 32. Kanchanatawan, B., S. Sirivichayakul, K. Ruxrungtham, A. F. Carvalho, M. Geffard, H. Ormstad, G. Anderson and M. Maes (2018). “Deficit, but Not Nondeficit, Schizophrenia Is Characterized by Mucosa-Associated Activation of the Tryptophan Catabolite (TRYCAT) Pathway with Highly Specific Increases in IgA Responses Directed to Picolinic, Xanthurenic, and Quinolinic Acid.” Mol Neurobiol 55(2): 1524–1536. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s12035-017-0417-6&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28181189&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) 33. Kim, H. S., S. Kim, S. J. Shin, Y. H. Park, Y. Nam, C. W. Kim, K. W. Lee, S. M. Kim, I. D. Jung, H. D. Yang, Y. M. Park and M. Moon (2021). “Gram-negative bacteria and their lipopolysaccharides in Alzheimer’s disease: pathologic roles and therapeutic implications.” Transl Neurodegener 10(1): 49. 34. LaFerla, F. M., K. N. Green and S. Oddo (2007). “Intracellular amyloid-beta in Alzheimer’s disease.” Nat Rev Neurosci 8(7): 499–509. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nrn2168&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17551515&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000247907800012&link_type=ISI) 35. Lugo-Huitrón, R., P. Ugalde Muñiz, B. Pineda, J. Pedraza-Chaverrí, C. Ríos and V. Pérez-de la Cruz (2013). “Quinolinic acid: an endogenous neurotoxin with multiple targets.” Oxid Med Cell Longev 2013: 104024. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1155/2013/104024&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24089628&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) 36. Maes, M., N. DeVos, A. Wauters, P. Demedts, V. Maurits, H. Neels, E. Bosmans, C. Altamura, A. Lin, C. Song, M. Vandenbroucke and S. Scharpe (1999). “Inflammatory markers in younger vs elderly normal volunteers and in patients with Alzheimer’s disease.” Journal of Psychiatric Research 33(5): 397–405. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0022-3956(99)00016-3&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=10504008&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000082296000005&link_type=ISI) 37. Maes, M., P. Galecki, Y. S. Chang and M. Berk (2011). “A review on the oxidative and nitrosative stress (O&NS) pathways in major depression and their possible contribution to the (neuro)degenerative processes in that illness.” Progress in Neuro-Psychopharmacology and Biological Psychiatry 35(3): 676–692. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.pnpbp.2010.05.004&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20471444&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) 38. Maes, M., B. E. Leonard, A. M. Myint, M. Kubera and R. Verkerk (2011). “The new ‘5-HT’ hypothesis of depression: cell-mediated immune activation induces indoleamine 2,3-dioxygenase, which leads to lower plasma tryptophan and an increased synthesis of detrimental tryptophan catabolites (TRYCATs), both of which contribute to the onset of depression.” Prog Neuropsychopharmacol Biol Psychiatry 35(3): 702–721. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.pnpbp.2010.12.017&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21185346&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) 39. Maes, M., B. E. Leonard, A. M. Myint, M. Kubera and R. Verkerk (2011). “The new ‘5-HT’ hypothesis of depression: Cell-mediated immune activation induces indoleamine 2,3-dioxygenase, which leads to lower plasma tryptophan and an increased synthesis of detrimental tryptophan catabolites (TRYCATs), both of which contribute to the onset of depression.” Progress in Neuro-Psychopharmacology and Biological Psychiatry 35(3): 702–721. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.pnpbp.2010.12.017&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21185346&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) 40. Maes, M., H. Y. Meltzer, S. Scharpé, E. Bosmans, E. Suy, I. De Meester, J. Calabrese and P. Cosyns (1993). “Relationships between lower plasma L-tryptophan levels and immune-inflammatory variables in depression.” Psychiatry Res 49(2): 151–165. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/0165-1781(93)90102-M&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=7908745&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1993MX09000005&link_type=ISI) 41. Mattson, M. P. (2004). “Pathways towards and away from Alzheimer’s disease.” Nature 430(7000): 631–639. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nature02621&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15295589&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000223085400034&link_type=ISI) 42. Moneim, A. E. A. (2015). “Oxidant/Antioxidant imbalance and the risk of Alzheimer’s disease.” Current Alzheimer research 12(4): 335–349. 43. Moroni, F. (1999). “Tryptophan metabolism and brain function: focus on kynurenine and other indole metabolites.” Eur J Pharmacol 375(1-3): 87–100. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0014-2999(99)00196-X&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=10443567&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000081483600009&link_type=ISI) 44. Morris, G., M. Berk, M. Maes and B. K. Puri (2019). “Could Alzheimer’s Disease Originate in the Periphery and If So How So?” Mol Neurobiol 56(1): 406–434. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s12035-018-1092-y&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=29705945&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) 45. Morris, G. P., I. A. Clark and B. Vissel (2014). “Inconsistencies and Controversies Surrounding the Amyloid Hypothesis of Alzheimer’s Disease.” Acta Neuropathologica Communications 2(1): 135. 46. O’Brien, R. J. and P. C. Wong (2011). “Amyloid precursor protein processing and Alzheimer’s disease.” Annu Rev Neurosci 34: 185–204. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1146/annurev-neuro-061010-113613&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21456963&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000293772100009&link_type=ISI) 47. Okuda, S., N. Nishiyama, H. Saito and H. Katsuki (1998). “3-Hydroxykynurenine, an endogenous oxidative stress generator, causes neuronal cell death with apoptotic features and region selectivity.” J Neurochem 70(1): 299–307. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=9422375&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000071146400035&link_type=ISI) 48. Oxenkrug, G., M. van der Hart, J. Roeser and P. Summergrad (2017). “Peripheral Tryptophan - Kynurenine Metabolism Associated with Metabolic Syndrome is Different in Parkinson’s and Alzheimer’s Diseases.” Endocrinology, diabetes and metabolism journal1(4): [http://researchopenworld.com/wp-content/uploads/2017/2011/EDMJ-2017-2113-Gregory-F-Oxenkrug-USA.pdf](http://researchopenworld.com/wp-content/uploads/2017/2011/EDMJ-2017-2113-Gregory-F-Oxenkrug-USA.pdf). 49. Oxenkrug, G. F., D. Gurevich, B. Siegel, M. S. Dumlao and S. Gershon (1989). “Correlation between brain-adrenal axis activation and cognitive impairment in alzheimer’s disease: Is there a gender effect?” Psychiatry Research 29(2): 169–175. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/0165-1781(89)90031-0&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=2798595&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) 50. Radulescu, I., A. M. Dragoi, S. C. Trifu and M. B. Cristea (2021). “Neuroplasticity and depression: Rewiring the brain’s networks through pharmacological therapy (Review).” Exp Ther Med 22(4): 1131. 51. Rahman, A., K. Ting, K. M. Cullen, N. Braidy, B. J. Brew and G. J. Guillemin (2009). “The excitotoxin quinolinic acid induces tau phosphorylation in human neurons.” PLoS One 4(7): e6344. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pone.0006344&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19623258&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) 52. Reddy, P. H. and M. F. Beal (2008). “Amyloid beta, mitochondrial dysfunction and synaptic damage: implications for cognitive decline in aging and Alzheimer’s disease.” Trends in Molecular Medicine 14(2): 45–53. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.molmed.2007.12.002&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=18218341&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000254233000001&link_type=ISI) 53. Rios, C. and A. Santamaria (1991). “Quinolinic acid is a potent lipid peroxidant in rat brain homogenates.” Neurochem Res 16(10): 1139–1143. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/BF00966592&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=1686636&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1991GK14800006&link_type=ISI) 54. Rommer, P. S., D. Fuchs, F. Leblhuber, R. Schroth, M. Greilberger, E. Tafeit and J. Greilberger (2016). “Lowered Levels of Carbonyl Proteins after Vitamin B Supplementation in Patients with Mild Cognitive Impairment and Alzheimer’s Disease.” Neurodegener Dis 16(3-4): 284–289. 55. Ruhé, H. G., N. S. Mason and A. H. Schene (2007). “Mood is indirectly related to serotonin, norepinephrine and dopamine levels in humans: a meta-analysis of monoamine depletion studies.” Mol Psychiatry 12(4): 331–359. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/sj.mp.4001949&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17389902&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000245313200005&link_type=ISI) 56. Santamaría, A., S. Galván-Arzate, V. Lisý, S. F. Ali, H. M. Duhart, L. Osorio-Rico, C. Ríos and F. St’astný (2001). “Quinolinic acid induces oxidative stress in rat brain synaptosomes.” Neuroreport 12(4): 871–874. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=11277599&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) 57. Schwarz, M. J., G. J. Guillemin, S. J. Teipel, K. Buerger and H. Hampel (2013). “Increased 3-hydroxykynurenine serum concentrations differentiate Alzheimer’s disease patients from controls.” Eur Arch Psychiatry Clin Neurosci 263(4): 345–352. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s00406-012-0384-x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23192697&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) 58. Smith, A. J., R. A. Smith and T. W. Stone (2009). “5-Hydroxyanthranilic acid, a tryptophan metabolite, generates oxidative stress and neuronal death via p38 activation in cultured cerebellar granule neurones.” Neurotox Res 15(4): 303–310. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19384564&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) 59. Smith, R. S. and M. Maes (1995). “The macrophage-T-lymphocyte theory of schizophrenia: additional evidence.” Med Hypotheses 45(2): 135–141. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/0306-9877(95)90062-4&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=8531836&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1995RW65600008&link_type=ISI) 60. Sorgdrager, F. J. H., Y. Vermeiren, M. Van Faassen, C. van der Ley, E. A. A. Nollen, I. P. Kema and P. P. De Deyn (2019). “Age- and disease-specific changes of the kynurenine pathway in Parkinson’s and Alzheimer’s disease.” J Neurochem 151(5): 656–668. 61. Tohgi, H., T. Abe, S. Takahashi, M. Kimura, J. Takahashi and T. Kikuchi (1992). “Concentrations of serotonin and its related substances in the cerebrospinal fluid in patients with Alzheimer type dementia.” Neuroscience Letters 141(1): 9–12. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/0304-3940(92)90322-X&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=1508406&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) 62. Tohgi, H., T. Abe, S. Takahashi, M. Saheki and M. Kimura (1995). “Indoleamine concentrations in cerebrospinal fluid from patients with Alzheimer type and Binswanger type dementias before and after administration of citalopram, a synthetic serotonin uptake inhibitor.” J Neural Transm Park Dis Dement Sect 9(2-3): 121–131. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/BF02259654&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=8526997&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1995RQ81700003&link_type=ISI) 63. van der Velpen, V., T. Teav, H. Gallart-Ayala, F. Mehl, I. Konz, C. Clark, A. Oikonomidi, G. Peyratout, H. Henry, M. Delorenzi, J. Ivanisevic and J. Popp (2019). “Systemic and central nervous system metabolic alterations in Alzheimer’s disease.” Alzheimers Res Ther 11(1): 93. 64. van der Wal, E. A., F. Gómez-Pinilla and C. W. Cotman (1993). “Transforming growth factor-beta 1 is in plaques in Alzheimer and Down pathologies.” Neuroreport 4(1): 69–72. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1097/00001756-199301000-00018&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=8453039&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1993KN46100018&link_type=ISI) 65. Vasupanrajit, A., K. Jirakran, C. Tunvirachaisakul and M. Maes (2021). “Suicide attempts are associated with activated immune-inflammatory, nitro-oxidative, and neurotoxic pathways: A systematic review and meta-analysis.” Journal of Affective Disorders 295: 80–92. 66. Vasupanrajit, A., K. Jirakran, C. Tunvirachaisakul, M. Solmi and M. Maes (2021). “Inflammation and nitro-oxidative stress in current suicidal attempts and current suicidal ideation: a systematic review and meta-analysis.” medRxiv: 2021.2009.2009.21263363. 67. Wennstrom, M., H. M. Nielsen, F. Orhan, E. Londos, L. Minthon and S. Erhardt (2014). “Kynurenic Acid levels in cerebrospinal fluid from patients with Alzheimer’s disease or dementia with lewy bodies.” Int J Tryptophan Res 7: 1–7. 68. Whiley, L., K. E. Chappell, E. D’Hondt, M. R. Lewis, B. Jimenez, S. G. Snowden, H. Soininen, I. Kloszewska, P. Mecocci, M. Tsolaki, B. Vellas, J. R. Swann, A. Hye, S. Lovestone, C. Legido-Quigley, E. Holmes and c. AddNeuroMed (2021). “Metabolic phenotyping reveals a reduction in the bioavailability of serotonin and kynurenine pathway metabolites in both the urine and serum of individuals living with Alzheimer’s disease.” Alzheimers Res Ther 13(1): 20. 69. Widner, B., F. Leblhuber, J. Walli, G. P. Tilz, U. Demel and D. Fuchs (2000). “Tryptophan degradation and immune activation in Alzheimer’s disease.” J Neural Transm (Vienna) 107(3): 343–353. 70. Willette, A. A., C. Pappas, N. Hoth, Q. Wang, B. Klinedinst, S. A. Willette, B. Larsen, A. Pollpeter, T. Li, S. Le, A. D. Collazo-Martinez, J. P. Mochel, K. Allenspach, R. Dantzer and I. Alzheimer’s Disease Neuroimaging (2021). “Inflammation, negative affect, and amyloid burden in Alzheimer’s disease: Insights from the kynurenine pathway.” Brain Behav Immun 95: 216–225. 71. Zadori, D., G. Veres, L. Szalardy, P. Klivenyi and L. Vecsei (2018). “Alzheimer’s Disease: Recent Concepts on the Relation of Mitochondrial Disturbances, Excitotoxicity, Neuroinflammation, and Kynurenines.” J Alzheimers Dis 62(2): 523–547. 72. Zhan, X., B. Stamova, L. W. Jin, C. DeCarli, B. Phinney and F. R. Sharp (2016). “Gram-negative bacterial molecules associate with Alzheimer disease pathology.” Neurology 87(22): 2324–2332. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1212/WNL.0000000000003391&link_type=DOI) 73. Zhang, R., R. G. Miller, R. Gascon, S. Champion, J. Katz, M. Lancero, A. Narvaez, R. Honrada, D. Ruvalcaba and M. S. McGrath (2009). “Circulating endotoxin and systemic immune activation in sporadic amyotrophic lateral sclerosis (sALS).” Journal of neuroimmunology 206(1-2): 121–124. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jneuroim.2008.09.017&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19013651&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F21%2F2022.03.18.22272608.atom) 74. Zhao, Y., V. Jaber and W. J. Lukiw (2017). “Secretory Products of the Human GI Tract Microbiome and Their Potential Impact on Alzheimer’s Disease (AD): Detection of Lipopolysaccharide (LPS) in AD Hippocampus.” Front Cell Infect Microbiol 7: 318. 75. Zvěřová, M., Z. Fišar, R. Jirák, E. Kitzlerová, J. Hroudová and J. Raboch (2013). “Plasma cortisol in Alzheimer’s disease with or without depressive symptoms.” Medical science monitor : international medical journal of experimental and clinical research 19: 681–689.