Ayşe Şiva Acar, Öykü Saday, Oytun Erbaş

Institute of Experimental Medicine, Kocaeli, Türkiye

Keywords: Autism spectrum disorder, depression, major depressive disorder, minocycline, schizophrenia, tetracycline.

Abstract

Tetracyclines are broad-spectrum antibiotics used primarily for bacterial infections and various other conditions. Experimental studies have demonstrated their neurogenesis-inducing, neuroprotective, antioxidant, antinociceptive, antiapoptotic, and anti-glutamate-induced excitotoxicity properties. Due to these attributes, it is believed that when combined with antipsychotic medications used in psychiatric disorders, tetracyclines may facilitate the treatment of conditions such as autism spectrum disorder (ASD), major depressive disorder (MDD), and schizophrenia. This review article examines the biochemical structure, pharmacokinetic, and pharmacodynamic properties of the tetracycline group of antibiotics, as well as their uses in various diseases. It particularly focuses on the neuroprotective, antioxidant, antinociceptive, antiapoptotic, and anti-glutamate-induced excitotoxicity properties of minocycline. Additionally, the potential use of minocycline in the treatment of psychiatric disorders such as ASD, MDD, and schizophrenia is discussed.

Introduction

Tetracyclines are a broad-spectrum group of antibiotics effective in inhibiting protein synthesis and are frequently used in the treatment of bacterial infections. This group includes therapeutics obtained naturally, such as tetracycline, oxytetracycline, and chlortetracycline, as well as semi-synthetic agents like minocycline (7-dimethylamino-6- demethyl-6-deoxytetracycline), doxycycline, and methacycline.[1]

Tetracycline antibiotics are indicated for a wide range of bacterial infections, including rickettsial infections, leptospirosis, amebiasis, actinomycosis, acne, nocardiosis, brucellosis, chlamydial infections, tularemia, Legionnaires' disease, syphilis, Lyme disease, and Whipple's disease. Beyond bacterial infections, they are also indicated for numerous other conditions such as rosacea, bullous dermatitis, Kaposi's sarcoma, sarcoidosis, pyoderma gangrenosum, hidradenitis suppurativa, alpha-1 antitrypsin deficiency, scleroderma, and rheumatoid arthritis.[1-4]

The tetracycline group is generally administered orally, but intramuscular, intravenous, and topical forms are also available. Only oxytetracycline and tetracycline can be used intramuscularly. This drug group has excellent tissue distribution, but its penetration into cerebrospinal fluid is poor. When taken with metals such as calcium, magnesium, or aluminum, chelation occurs, which reduces absorption.[5,6]

The use of tetracycline can cause side effects such as discoloration of the teeth, epigastric pain, nausea, vomiting, anorexia, photosensitivity, delayed bone development in children, and Clostridium difficile infection, which is a common side effect seen with many antibiotics.[7,8] Additionally, tetracyclines are contraindicated in pregnant women due to their potential to cause discoloration of the fetus's teeth, impairment of fetal long bone growth, and hepatotoxicity.[9-11] Tetracyclines also pass into breast milk; however, the calcium present in breast milk chelates the drug, preventing its transfer to the infant. Therefore, tetracyclines are considered safe therapeutic agents for breastfeeding mothers and their infants.[12]

Minocycline is a second-generation, semi-synthetic, potent tetracycline analog with neuroprotective and anti-inflammatory effects.[13] Its lipophilic structure allows it to cross the blood-brain barrier and be readily absorbed by the body.[14] Although its immunomodulatory mechanism has not been fully elucidated, minocycline affects multiple pathways.[15] It reduces microglial activation,[16] inhibits matrix metalloproteinase-9 activity, and modulates the secretion of cytokines and chemokines involved in neuroinflammation, such as interleukin-6 and tumor necrosis factor-alpha (TNF-α).[17]

Minocycline increases the expression of the anti-apoptotic B cell lymphoma 2 (Bcl-2) and reduces the expression of pro-apoptotic proteins such as Bax and Bid, thereby balancing the Bax/Bcl-2 ratio.[18] This results in the suppression of apoptotic factors such as caspase-1 and caspase-3 activation and cytochrome c.[19,20]

Although minocycline is actively used in the treatment of acne vulgaris or as an antiviral agent, its anti-inflammatory, antioxidant, and antiapoptotic effects have led scientists to investigate its potential beyond these usage targets. Research has been directed towards exploring its potential in neuro-psychiatric conditions such as Alzheimer’s, Parkinson’s, and Huntington's diseases.[21-28]

Minocycline, as an inhibitor of microglial activation, possesses anti-inflammatory, antioxidant, and antiapoptotic properties, and it can modulate glutamate-dependent excitotoxicity. Its therapeutic potential has been demonstrated through various animal experiments.[28-35] In addition to these findings, recent experiments have shown that minocycline, when used in combination with antipsychotic medications, particularly improves negative symptoms.[36-41]

THE POTENTIAL ANTIDEPRESSANT EFFECTS OF MINOCYCLINE

Depression treatment

Both clinical and preclinical studies indicate that major depressive disorder (MDD) stems from inadequate inflammatory responses and impaired neuroprotection. Researchers suggest that minocycline, a second-generation tetracycline antibiotic, could play a role in the treatment of major depression by virtue of its identified antinociceptive effects. Moreover, it is proposed that minocycline could be utilized in the treatment of both MDD and somatoform disorders, as well as certain prominent somatic disorders. Results from various studies conducted on humans and animals also indicate that minocycline possesses antidepressant-like and anti-inflammatory effects on the nervous system. Its antidepressant properties have also been evidenced through the ‘forced swim test’.[42]

In light of recent research and findings, the pathogenesis of MDD has been expanded to encompass limbic system damage and alterations in inflammatory mediators in the brain. This includes disrupted neuroprotection. Examples of this include the repeated findings of decreased neuronal survival and impaired neurogenesis in the hippocampus in patients with MDD. This situation strongly supports the consideration of the aforementioned findings as possible elements of the disorder's pathophysiology.[43,44]

Another characteristic of minocycline is its ability to effectively cross the blood-brain barrier. Neuroprotective effects of minocycline have been observed in clinical and preclinical studies in amyotrophic lateral sclerosis and Parkinson's disease.[45,46] In animal experiments, it has been understood that minocycline significantly increases neurogenesis and markedly reduces 3,4-methylenedioxymethamphetamine-induced serotonin and dopamine system neurotoxicity in the cerebral cortex and hippocampus.[46] Minocycline has a regulatory effect on proinflammatory agents such as TNF-α, nitric oxide, and interleukin-1 beta.[47] Additionally, it has been reported that these agents increase in patients with MDD and return to normal levels after antidepressant treatment.[43,48] It is also suggested that minocycline may exert potential antidepressant effects through its strong neuroprotective activities, which include neurogenesis, antioxidation, anti-glutamateinduces excitotoxicity, and direct regulation of proinflammatory agents.[42]

Neurogenesis, antioxidation, and anti-glutamate-induced excitotoxicity

The hippocampus, a crucial part of the limbic system, is predominantly affected in depression and regulates activities such as mood, anxiety, learning, and memory.[47] Clinical and preclinical studies indicate that another consequence of MDD is a reduction in the total number of neurons in the hippocampus, mainly due to a prolonged decrease in cell proliferation rate.[49] In contrast, antidepressant treatments accelerate hippocampal neurogenesis; thus, they can prevent or reverse the atrophy and damage caused by major depression. These mechanisms have been demonstrated with fluoxetine, tianeptine, and lithium.[50-52] In light of these findings, it is assumed that the secondgeneration tetracycline antibiotic minocycline may directly attenuate or reverse neuronal atrophy caused by MDD by up-regulating hippocampal neurogenesis.[42]

Levels of lipid peroxidation and oxidative damage markers such as superoxide dismutase are observed to increase in the serum of major depression patients, while antioxidants such as vitamins E and C decrease. However, after effective antidepressant treatment, these levels return to normal.[53,54] It has been shown that minocycline possesses a direct radical scavenging property similar to vitamin E, and upon examination of the drug's chemical structure, it becomes apparent that minocycline contains multiple phenolic rings similar to vitamin E.[55]

Plasma and frontal cortex glutamate levels have been found to be higher in patients with MDD compared to the normal control group.[55,56] Antidepressant treatment, on the other hand, significantly increases glutamate levels.[57] In recent times, it has been understood that anti-glutamatergic agents also have antidepressant effects.[58] Therefore, the direct and potent neuroprotective effect of minocycline against glutamate excitotoxicity increases the possibility of another antidepressant mechanism through the regulation of the p38 and Akt pathways.[59]

The proven antidepressant effect of minocycline with the forced swim test

Minocycline has demonstrated antidepressant-like actions by increasing climbing behavior, reducing immobility, and synergizing with subthreshold doses of desipramine and glutamate receptor antagonists. Molina-Hernández et al.[60] suggested that the antidepressant effect of minocycline may involve modification of the noradrenergic system in the brain. Interestingly, minocycline did not synergize with fluoxetine's antidepressant-like effects, indicating that minocycline may not directly affect the serotonergic system. In light of this information, it can be said that minocycline may be used to enhance the effect of noradrenergic antidepressant drugs in this regard.

Minocycline has been shown to have neuroprotective effects in various regions of the brain, and it is thought that it may be used not only in older patients with MDD but also in comorbid conditions associated with organic brain pathologies such as Parkinson's disease, stroke, traumatic brain injury, Alzheimer's, and others.[61] It has also been shown that minocycline has a strong and consistent antinociceptive effect, demonstrated in pain models associated with tissue damage and inflammation. This suggests that minocycline has been effective in treating somatic symptoms commonly found in patients with major depression and pain disorders such as fibromyalgia, where functional somatoform disorders are often present.[62] According to the results of another study, the addition of minocycline to clomipramine (150 mg/day) has led to significant improvements in depressive symptoms within 3-4 days, with the Hamilton Depression Rating Scale score decreasing from 25 to 8. It was reported that this effect continued after minocycline was discontinued (after a 2-week treatment) and was also associated with the resolution of facial pain.[63] Briefly, this study demonstrates that minocycline can be highly beneficial in producing antidepressant effects when used in conjunction with a serotonergic antidepressant to enhance its effects. However, this information contradicts the synergy study conducted with desipramine, a noradrenergic antidepressant, rather than fluoxetine, a serotonergic antidepressant agent used in the forced swim test animal model.[60]

Tetracycline use in the treatment of schizophrenia

Schizophrenia is a psychiatric disorder characterized by delusions, hallucinations, disorganized speech, decreased motivation, and cognitive impairments. Affecting approximately 1% of the world population, this illness ranks among the top 10 leading causes of global disability.[64]

The results of studies so far suggest that neuroinflammation affecting microglial cells plays a significant role in the etiology of schizophrenia, and appropriately controlling microglial activation holds promise in the therapeutic strategy for schizophrenia.[65,66]

The second-generation tetracycline antibiotic minocycline, which inhibits microglial activation, has shown neuroprotective effects in many different neurodegenerative diseases. Mechanisms overlapping with neuropathological pathways are being investigated for minocycline's role as an adjunctive treatment in schizophrenia and its potential use in treatment for its ability to improve negative symptoms of the disease.[67]

Until now, the effective treatment of schizophrenia has been limited to antipsychotic medications with antidopaminergic effects, which regulate dysfunctional neurotransmitter systems to alleviate symptoms of the disease.[68] Additionally, while antipsychotic medications are the most effective drug class for positive symptoms, they are not sufficiently effective for negative and cognitive symptoms.[69] In recent years, the results of studies have strengthened the concept of microglial neuroinflammation in etiology. Particularly, the appropriate control of microglial activation holds great promise for the treatment of the disease.[70-73]

Minocycline has a distinct neuroprotective profile independent of its antibacterial properties.[13] Almost all of it is absorbed when taken orally, and it penetrates the brain tissue very well. These properties, along with the beneficial effects observed in animal experiments related to neurological disorders, have prompted scientists to investigate the potential role of the drug in the treatment of schizophrenia.[13,28] Subsequent studies have reported many beneficial effects of minocycline in treatment when used in combination with antipsychotic medications.[30,38,74,75]

Preclinical studies

Preclinical studies indicate the potential neurotherapeutic effects of minocycline in mental illnesses.[28-31] Another reported finding is that similar to its benefits in dopaminergic terminal neurotoxicity, minocycline can also induce behavioral changes related to hallucinations and delusions.[29]

According to Hashimoto's animal experiments with monkeys, minocycline protects the brain from methamphetamine-induced neurotoxicity in the monkey brain.[32] In Levkovitz et al.’s[30] study, the effects of minocycline and haloperidol on schizophrenia were compared in animal experiments. Rats were administered minocycline at a dose of 35 mg/kg/day for three days, followed by injection of MK-801 (N-methylD-aspartate receptor agonist), and evaluated using behavioral tests. The findings showed that MK-801 caused cognitive visual-spatial memory impairments and significantly disrupted sensorimotor gating, similar to schizophrenia, and minocycline reversed these cognitive impairments like haloperidol. Similarly, Zhang et al.[33] suggested that minocycline might have a potential therapeutic role in schizophrenia by reducing behavioral abnormalities like acute hyperactivity in mice following dizocilpine administration.

Clinical studies

Interestingly, some studies have shown that minocycline has significant effects on negative symptoms and contributes to impaired cognitive function and weakened vocational and social skills in schizophrenia patients. Case reports demonstrate the effective role of minocycline when added to antipsychotic medications for treating negative symptoms of schizophrenia.[36,37]

According to a randomized, double-blind, placebo-controlled clinical trial, the addition of minocycline (200 mg/day) to the treatment of patients with early psychosis for one year was reported to reduce negative symptoms of schizophrenia without detectable adverse effects on cognition.[38]

Sofuo¤lu et al.[76] demonstrated in their study that minocycline improved subjective reward effects in dextroamphetamine-dependent individuals.

Tetracyclines in Autism

Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by symptoms such as difficulties in socialization and highly repetitive patterns of behavior.[77] The prevalence of the disorder, which is observed in 1% of the world's population, is rapidly increasing day by day.[78,79] Although many environmental factors such as socioeconomic status, race, and culture influence the etiology of the disease, genetic factors are among the most important factors in the etiology of ASD.[80,81] Autism is more prevalent in males than in females, and comorbid conditions are also more frequently observed in males (70% concurrent conditions).[82] In addition, large-effect rare mutations or numerous small-effect mutations also play a significant role in the etiology of the disease.[77]

Individuals with ASD have atypical social perceptions, executive dysfunctions, and unusual cognitive behaviors, which are believed to be associated with abnormal neuronal development.[83] Due to these behaviors, individuals with ASD can be easily distinguished within social communities. Although ASD can be diagnosed easily in the preschool years, early diagnosis and intervention are extremely important for the patient's quality of life. Evaluations should be multidisciplinary and transformative in light of the Diagnostic and Statistical Manual of Mental Disorders-5 diagnostic criteria.[84] Following an early diagnosis, interventions aimed at treatment goals can enhance social communication and reduce aggressive behaviors. Although therapeutic agents prescribed after diagnosis can alleviate comorbid symptoms, being in a supportive environment that respects the differences of individuals with ASD and makes them feel safe is also extremely valuable for their quality of life.[85]

Studies conducted with minocycline have primarily focused on Fragile X syndrome, an X-chromosome-linked disorder, which is one of the most common causes of ASD.[86,87] In Fragile X syndrome, mutations occurring in the Fragile X mental retardation 1 (FMR1) gene on the X chromosome result in cognitive dysfunction, changes in visual and auditory capabilities, hyperactivity, communication problems, and ASD in affected individuals.[88,89] In an open-label study, it has been demonstrated that minocycline significantly increases sensitivity in Fragile X syndrome patients, as measured by the Aberrant Behavior Checklist-Community (ABC-C) sensitivity subscale.[90] In another study, it was observed that dendritic maturation increased with minocycline, and the performance of FMR1 genesilenced mice decreased.[86] Based on clinical trials conducted with individuals with Fragile X syndrome, minocycline has been observed to be a safe therapeutic agent for improving autistic behaviors.[91-93]

After administering minocycline (1.4 mg/kg/day) for six months to 11 autistic children, along with vitamin B6 (0.6 mg/kg) treatment to reduce vestibular side effects, the experiment was completed with 10 children. Among them, while the severity scores remained stable on the Clinical Global Impression (CGI) scale for eight children, only minimal improvement was observed in the severity score of CGI for two children, and clinical improvements remained negligible. The composite scores of the Vineland Adaptive Behavior Scale showed very little change, and several side effects were reported by parents. While a significant decrease in cytokine release was observed only in interleukin-8, there was no increase in colony-stimulating factor.[94]

In a randomized, double-blind, placebo-controlled study conducted over 10 weeks with 50 children with ASD, the administration of minocycline in addition to the most prescribed atypical antipsychotic, risperidone, resulted in a significant decrease in sensitivity, hyperactivity, and irritability subscales of the ABC-C. Based on these results, minocycline has been reported as an effective therapeutic target in the treatment of ASD.[95]

In a study conducted using mouse models of autism, it was observed that adding minocycline to the drinking water from the 17th day of pregnancy to the 21st day after birth resulted in improvement in microglial activation and anxiety behaviors.[96] Another study conducted with a rat model of autism induced by valproic acid also showed that minocycline use improved autistic behaviors.[92]

After 10 days of minocycline treatment (oral gavage, 30 mg/kg per day) given to adult mice with silenced FMR1 gene, it was observed that the treatment increased gamma-band phase-locking in response to auditory stimuli and decreased gamma power in resting state electroencephalogram.[93]

In conclusion, in light of the studies conducted, especially with minocycline, this broad-spectrum antibiotic group, which is used in many diseases besides its primary therapeutic target of bacterial diseases, is now seen as promising for psychiatric disorders as well. In addition to its known neuroprotective and anti-inflammatory effects, minocycline, which has been shown to have antidepressant effects in vivo and in vitro animal experiments, can be suggested to enhance the effects of noradrenergic antidepressant drugs in the treatment of MDD. For ASD, more extensive studies across age groups are needed to demonstrate the effects of the drug in different age groups. Furthermore, research should be intensified on the positive effects of the combination of risperidone and minocycline. The use of minocycline in the treatment of schizophrenia is recommended by scientists based on studies and meta-analyses. Clinical trials do not provide definitive information due to high dropout rates, but preclinical animal experiments provide promising results for the use of tetracycline group antibiotics as a new alternative in the treatment of psychiatric disorders.

Cite this article as: Şiva Acar A, Saday Ö, Erbaş O. Tetracycline antibiotics: Emerging roles in neuropsychiatric disorders. D J Med Sci 2024;10(2):74-82. doi: 10.5606/fng. btd.2024.149.

Author Contributions

All authors contributed equallyto this article.

Conflict of Interest

The authors declared no conflicts of interest with respect to the authorship and/or publication of this article.

Financial Disclosure

The authors received no financial support for the research and/or authorship of this article.

Data Sharing Statement:
The data that support the findings of this study are available from the corresponding author upon reasonable request.

References

  1. Pallett AP, Smyth EG. Clinicians' guide to antibiotics. Tetracycline. Br J Hosp Med 1988;40:385-90.
  2. Sapadin AN, Fleischmajer R. Tetracyclines: Nonantibiotic properties and their clinical implications. J Am Acad Dermatol 2006;54:258-65. doi: 10.1016/j. jaad.2005.10.004.
  3. Nelson ML, Levy SB. The history of the tetracyclines. Ann N Y Acad Sci 2011;1241:17-32. doi: 10.1111/j.1749- 6632.2011.06354.x.
  4. Dokuyucu R, Kokacya H, Inanir S, Copoglu US, Erbas O. Antipsychotic-like effect of minocycline in a rat model. Int J Clin Exp Med 2014;7:3354-61.
  5. Dougherty JA, Sucher AJ, Chahine EB, Shihadeh KC. Omadacycline: A new tetracycline antibiotic. Ann Pharmacother 2019;53:486-500. doi: 10.1177/1060028018818094.
  6. Bernier C, Dréno B. Minocycline. Ann Dermatol Venereol 2001;128:627-37.
  7. Sánchez AR, Rogers RS 3rd, Sheridan PJ. Tetracycline and other tetracycline-derivative staining of the teeth and oral cavity. Int J Dermatol 2004;43:709-15. doi: 10.1111/j.1365-4632.2004.02108.x.
  8. Demers P, Fraser D, Goldbloom RB, Haworth JC, LaRochelle J, MacLean R, et al. Effects of tetracyclines on skeletal growth and dentition. A report by the Nutrition Committee of the Canadian Paediatric Society. Can Med Assoc J 1968;99:849-54.
  9. Sukhorukikh SV. Tetracyclines in pregnancy. Br Med J 1965;1:743-4.
  10. Cross R, Ling C, Day NP, McGready R, Paris DH. Revisiting doxycycline in pregnancy and early childhood--time to rebuild its reputation? Expert Opin Drug Saf 2016;15:367- 82. doi: 10.1517/14740338.2016.1133584.
  11. Heaton PC, Fenwick SR, Brewer DE. Association between tetracycline or doxycycline and hepatotoxicity: A population based case-control study. J Clin Pharm Ther 2007;32:483-7. doi: 10.1111/j.1365- 2710.2007.00853.x.
  12. Chung AM, Reed MD, Blumer JL. Antibiotics and breastfeeding: A critical review of the literature. Paediatr Drugs 2002;4:817-37. doi: 10.2165/00128072-200204120- 00006.
  13. Yong VW, Wells J, Giuliani F, Casha S, Power C, Metz LM. The promise of minocycline in neurology. Lancet Neurol 2004;3:744-51. doi: 10.1016/S1474- 4422(04)00937-8.
  14. Chopra I, Roberts M. Tetracycline antibiotics: Mode of action, applications, molecular biology, and epidemiology of bacterial resistance. Microbiol Mol Biol Rev 2001;65:232-60. doi: 10.1128/MMBR.65.2.232- 260.2001.
  15. Kim HS, Suh YH. Minocycline and neurodegenerative diseases. Behav Brain Res 2009;196:168-79. doi: 10.1016/j.bbr.2008.09.040.
  16. Zhu F, Zheng Y, Ding YQ, Liu Y, Zhang X, Wu R, et al. Minocycline and risperidone prevent microglia activation and rescue behavioral deficits induced by neonatal intrahippocampal injection of lipopolysaccharide in rats. PLoS One 2014;9:e93966. doi: 10.1371/journal. pone.0093966.
  17. Siller SS, Broadie K. Matrix metalloproteinases and minocycline: Therapeutic avenues for fragile X syndrome. Neural Plast 2012;2012:124548. doi: 10.1155/2012/124548.
  18. Sinha-Hikim I, Shen R, Nzenwa I, Gelfand R, Mahata SK, Sinha-Hikim AP. Minocycline suppresses oxidative stress and attenuates fetal cardiac myocyte apoptosis triggered by in utero cocaine exposure. Apoptosis 2011;16:563-73. doi: 10.1007/s10495-011-0590-4.
  19. Zhang L, Huang P, Chen H, Tan W, Lu J, Liu W, et al. The inhibitory effect of minocycline on radiation-induced neuronal apoptosis via AMPKa1 signaling-mediated autophagy. Sci Rep 2017;7:16373. doi: 10.1038/ s41598-017-16693-8.
  20. Castanares M, Vera Y, Erkkilä K, Kyttänen S, Lue Y, Dunkel L, et al. Minocycline up-regulates BCL-2 levels in mitochondria and attenuates male germ cell apoptosis. Biochem Biophys Res Commun 2005;337:663-9. doi: 10.1016/j.bbrc.2005.09.101.
  21. Garner SE, Eady A, Bennett C, Newton JN, Thomas K, Popescu CM. Minocycline for acne vulgaris: Efficacy and safety. Cochrane Database Syst Rev 2012;2012:CD002086. doi: 10.1002/14651858. CD002086.pub2.
  22. Nagarakanti S, Bishburg E. Is minocycline an antiviral agent? A review of current literature. Basic Clin Pharmacol Toxicol 2016;118:4-8. doi: 10.1111/bcpt.12444.
  23. Clemens V, Regen F, Le Bret N, Heuser I, HellmannRegen J. Anti-inflammatory effects of minocycline are mediated by retinoid signaling. BMC Neurosci 2018;19:58. doi: 10.1186/s12868-018-0460-x.
  24. Regen F, Le Bret N, Hildebrand M, Herzog I, Heuser I, Hellmann-Regen J. Inhibition of brain retinoic acid catabolism: A mechanism for minocycline's pleiotropic actions? World J Biol Psychiatry 2016;17:634-40. doi: 10.3109/15622975.2015.1036116.
  25. Choi Y, Kim HS, Shin KY, Kim EM, Kim M, Kim HS, et al. Minocycline attenuates neuronal cell death and improves cognitive impairment in Alzheimer's disease models. Neuropsychopharmacology 2007;32:2393- 404. doi: 10.1038/sj.npp.1301377.
  26. Abdel-Salam OM. Drugs used to treat Parkinson's disease, present status and future directions. CNS Neurol Disord Drug Targets 2008;7:321-42. doi: 10.2174/187152708786441867.
  27. Chen M, Ona VO, Li M, Ferrante RJ, Fink KB, Zhu S, et al. Minocycline inhibits caspase-1 and caspase-3 expression and delays mortality in a transgenic mouse model of Huntington disease. Nat Med 2000;6:797-801. doi: 10.1038/77528.
  28. Dean OM, Data-Franco J, Giorlando F, Berk M. Minocycline: Therapeutic potential in psychiatry. CNS Drugs 2012;26:391-401. doi: 10.2165/11632000- 000000000-00000.
  29. Zhang L, Kitaichi K, Fujimoto Y, Nakayama H, Shimizu E, Iyo M, et al. Protective effects of minocycline on behavioral changes and neurotoxicity in mice after administration of methamphetamine. Prog Neuropsychopharmacol Biol Psychiatry 2006;30:1381-93. doi: 10.1016/j. pnpbp.2006.05.015.
  30. Levkovitz Y, Levi U, Braw Y, Cohen H. Minocycline, a second-generation tetracycline, as a neuroprotective agent in an animal model of schizophrenia. Brain Res 2007;1154:154-62. doi: 10.1016/j. brainres.2007.03.080.
  31. Monte AS, de Souza GC, McIntyre RS, Soczynska JK, dos Santos JV, Cordeiro RC, et al. Prevention and reversal of ketamine-induced schizophrenia related behavior by minocycline in mice: Possible involvement of antioxidant and nitrergic pathways. J Psychopharmacol 2013;27:1032-43. doi: 10.1177/0269881113503506.
  32. Hashimoto K, Tsukada H, Nishiyama S, Fukumoto D, Kakiuchi T, Iyo M. Protective effects of minocycline on the reduction of dopamine transporters in the striatum after administration of methamphetamine: A positron emission tomography study in conscious monkeys. Biol Psychiatry 2007;61:577-81. doi: 10.1016/j. biopsych.2006.03.019.
  33. Zhang L, Shirayama Y, Iyo M, Hashimoto K. Minocycline attenuates hyperlocomotion and prepulse inhibition deficits in mice after administration of the NMDA receptor antagonist dizocilpine. Neuropsychopharmacology 2007;32:2004-10. doi: 10.1038/sj.npp.1301313.
  34. Kunitachi S, Fujita Y, Ishima T, Kohno M, Horio M, Tanibuchi Y, et al. Phencyclidine-induced cognitive deficits in mice are ameliorated by subsequent subchronic administration of donepezil: Role of sigma-1 receptors. Brain Res 2009;1279:189-96. doi: 10.1016/j. brainres.2009.05.004.
  35. Mizoguchi H, Takuma K, Fukakusa A, Ito Y, Nakatani A, Ibi D, et al. Improvement by minocycline of methamphetamine-induced impairment of recognition memory in mice. Psychopharmacology (Berl) 2008;196:233-41. doi: 10.1007/s00213-007-0955-0.
  36. Kelly DL, Vyas G, Richardson CM, Koola M, McMahon RP, Buchanan RW, et al. Adjunct minocycline to clozapine treated patients with persistent schizophrenia symptoms. Schizophr Res 2011;133:257-8. doi: 10.1016/j.schres.2011.08.005.
  37. Jhamnani K, Shivakumar V, Kalmady S, Rao NP, Venkatasubramanian G. Successful use of addon minocycline for treatment of persistent negative symptoms in schizophrenia. J Neuropsychiatry Clin Neurosci 2013;25:E06-7. doi: 10.1176/appi. neuropsych.11120376.
  38. Chaudhry IB, Hallak J, Husain N, Minhas F, Stirling J, Richardson P, et al. Minocycline benefits negative symptoms in early schizophrenia: A randomised doubleblind placebo-controlled clinical trial in patients on standard treatment. J Psychopharmacol 2012;26:1185- 93. doi: 10.1177/0269881112444941.
  39. Miyaoka T, Yasukawa R, Yasuda H, Hayashida M, Inagaki T, Horiguchi J. Possible antipsychotic effects of minocycline in patients with schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 2007;31:304-7. doi: 10.1016/j.pnpbp.2006.08.013.
  40. Chaves C, de Marque CR, Wichert-Ana L, Maia-deOliveira JP, Itikawa EN, Crippa JA, et al. Functional neuroimaging of minocycline's effect in a patient with schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 2010;34:550-2. doi: 10.1016/j. pnpbp.2010.01.020.
  41. Miyaoka T, Yasukawa R, Yasuda H, Hayashida M, Inagaki T, Horiguchi J. Minocycline as adjunctive therapy for schizophrenia: An open-label study. Clin Neuropharmacol 2008;31:287-92. doi: 10.1097/ WNF.0b013e3181593d45.
  42. Pae CU, Marks DM, Han C, Patkar AA. Does minocycline have antidepressant effect? Biomed Pharmacother 2008;62:308-11. doi: 10.1016/j.biopha.2007.12.005.
  43. Maes M. Evidence for an immune response in major depression: A review and hypothesis. Prog Neuropsychopharmacol Biol Psychiatry 1995;19:11-38. doi: 10.1016/0278-5846(94)00101-m.
  44. Loftis JM, Huckans M, Morasco BJ. Neuroimmune mechanisms of cytokine-induced depression: Current theories and novel treatment strategies. Neurobiol Dis 2010;37:519-33. doi: 10.1016/j.nbd.2009.11.015.
  45. Traynor BJ, Bruijn L, Conwit R, Beal F, O'Neill G, Fagan SC, et al. Neuroprotective agents for clinical trials in ALS: A systematic assessment. Neurology 2006;67:20- 7. doi: 10.1212/01.wnl.0000223353.34006.54.
  46. NINDS NET-PD Investigators. A randomized, doubleblind, futility clinical trial of creatine and minocycline in early Parkinson disease. Neurology 2006;66:664- 71. doi: 10.1212/01.wnl.0000201252.57661.e1.
  47. Pala HG, Erbas O, Pala EE, Artunc Ulkumen B, Akman L, Akman T, Oltulu F, Yavasoglu A. The effects of sunitinib on endometriosis. J Obstet Gynaecol. 2015 Feb;35(2):183-7. doi: 10.3109/01443615.2014.941345. Epub 2014 Aug 5. PMID: 25093747.
  48. Pala HG, Erbas O, Pala EE, Artunc Ulkumen B, Akman L, Akman T, et al. The effects of sunitinib on endometriosis. J Obstet Gynaecol 2015;35:183-7. doi: 10.3109/01443615.2014.941345.
  49. Herken H, Gurel A, Selek S, Armutcu F, Ozen ME, Bulut M, et al. Adenosine deaminase, nitric oxide, superoxide dismutase, and xanthine oxidase in patients with major depression: Impact of antidepressant treatment. Arch Med Res 2007;38:247-52. doi: 10.1016/j. arcmed.2006.10.005.
  50. Warner-Schmidt JL, Duman RS. Hippocampal neurogenesis: Opposing effects of stress and antidepressant treatment. Hippocampus 2006;16:239- 49. doi: 10.1002/hipo.20156.
  51. Chiou SH, Chen SJ, Peng CH, Chang YL, Ku HH, Hsu WM, et al. Fluoxetine up-regulates expression of cellular FLICE-inhibitory protein and inhibits LPSinduced apoptosis in hippocampus-derived neural stem cell. Biochem Biophys Res Commun 2006;343:391- 400. doi: 10.1016/j.bbrc.2006.02.180.
  52. Czéh B, Michaelis T, Watanabe T, Frahm J, de Biurrun G, van Kampen M, et al. Stress-induced changes in cerebral metabolites, hippocampal volume, and cell proliferation are prevented by antidepressant treatment with tianeptine. Proc Natl Acad Sci U S A 2001;98:12796-801. doi: 10.1073/pnas.211427898.
  53. Manji HK, Moore GJ, Chen G. Clinical and preclinical evidence for the neurotrophic effects of mood stabilizers: Implications for the pathophysiology and treatment of manic-depressive illness. Biol Psychiatry 2000;48:740- 54. doi: 10.1016/s0006-3223(00)00979-3.
  54. Bilici M, Efe H, Köroğlu MA, Uydu HA, Bekaroğlu M, Değer O. Antioxidative enzyme activities and lipid peroxidation in major depression: Alterations by antidepressant treatments. J Affect Disord 2001;64:43- 51. doi: 10.1016/s0165-0327(00)00199-3.
  55. Maes M, De Vos N, Pioli R, Demedts P, Wauters A, Neels H, et al. Lower serum vitamin E concentrations in major depression. Another marker of lowered antioxidant defenses in that illness. J Affect Disord 2000;58:241-6. doi: 10.1016/s0165-0327(99)00121-4.
  56. Hashimoto K, Sawa A, Iyo M. Increased levels of glutamate in brains from patients with mood disorders. Biol Psychiatry 2007;62:1310-6. doi: 10.1016/j. biopsych.2007.03.017.
  57. Mauri MC, Ferrara A, Boscati L, Bravin S, Zamberlan F, Alecci M, et al. Plasma and platelet amino acid concentrations in patients affected by major depression and under fluvoxamine treatment. Neuropsychobiology 1998;37:124-9. doi: 10.1159/000026491.
  58. Maes M, Verkerk R, Vandoolaeghe E, Lin A, Scharpé S. Serum levels of excitatory amino acids, serine, glycine, histidine, threonine, taurine, alanine and arginine in treatment-resistant depression: Modulation by treatment with antidepressants and prediction of clinical responsivity. Acta Psychiatr Scand 1998;97:302- 8. doi: 10.1111/j.1600-0447.1998.tb10004.x.
  59. Coric V, Milanovic S, Wasylink S, Patel P, Malison R, Krystal JH. Beneficial effects of the antiglutamatergic agent riluzole in a patient diagnosed with obsessivecompulsive disorder and major depressive disorder. Psychopharmacology (Berl) 2003;167:219-20. doi: 10.1007/s00213-003-1396-z.
  60. Pi R, Li W, Lee NT, Chan HH, Pu Y, Chan LN, et al. Minocycline prevents glutamate-induced apoptosis of cerebellar granule neurons by differential regulation of p38 and Akt pathways. J Neurochem 2004;91:1219- 30. doi: 10.1111/j.1471-4159.2004.02796.x.
  61. Molina-Hernández M, Tellez-Alcántara NP, Pérez-García J, Olivera-Lopez JI, Jaramillo-Jaimes MT. Antidepressantlike actions of minocycline combined with several glutamate antagonists. Prog Neuropsychopharmacol Biol Psychiatry 2008;32:380-6. doi: 10.1016/j. pnpbp.2007.09.004.
  62. Hunter CL, Quintero EM, Gilstrap L, Bhat NR, Granholm AC. Minocycline protects basal forebrain cholinergic neurons from mu p75-saporin immunotoxic lesioning. Eur J Neurosci 2004;19:3305-16. doi: 10.1111/j.0953- 816X.2004.03439.x.
  63. Hua XY, Svensson CI, Matsui T, Fitzsimmons B, Yaksh TL, Webb M. Intrathecal minocycline attenuates peripheral inflammation-induced hyperalgesia by inhibiting p38 MAPK in spinal microglia. Eur J Neurosci 2005;22:2431-40. doi: 10.1111/j.1460- 9568.2005.04451.x.
  64. Levine J, Cholestoy A, Zimmerman J. Possible antidepressant effect of minocycline. Am J Psychiatry 1996;153:582. doi: 10.1176/ajp.153.4.582b.
  65. Fleischhacker WW, Arango C, Arteel P, Barnes TR, Carpenter W, Duckworth K, et al. Schizophrenia--time to commit to policy change. Schizophr Bull 2014;40 Suppl 3:S165-94. doi: 10.1093/schbul/sbu006.
  66. Zheng W, Zhu XM, Zhang QE, Cheng G, Cai DB, He J, et al. Adjunctive minocycline for major mental disorders: A systematic review. J Psychopharmacol 2019;33:1215-26. doi: 10.1177/0269881119858286.
  67. Solmi M, Veronese N, Thapa N, Facchini S, Stubbs B, Fornaro M, et al. Systematic review and metaanalysis of the efficacy and safety of minocycline in schizophrenia. CNS Spectr 2017;22:415-26. doi: 10.1017/S1092852916000638.
  68. Zhang L, Zhao J. Profile of minocycline and its potential in the treatment of schizophrenia. Neuropsychiatr Dis Treat 2014;10:1103-11. doi: 10.2147/NDT.S64236.
  69. Kane JM, Correll CU. Past and present progress in the pharmacologic treatment of schizophrenia. J Clin Psychiatry 2010;71:1115-24. doi: 10.4088/ JCP.10r06264yel.
  70. Leucht S, Arbter D, Engel RR, Kissling W, Davis JM. How effective are second-generation antipsychotic drugs? A meta-analysis of placebo-controlled trials. Mol Psychiatry 2009;14:429-47. doi: 10.1038/sj.mp.4002136.
  71. Monji A, Kato T, Kanba S. Cytokines and schizophrenia: Microglia hypothesis of schizophrenia. Psychiatry Clin Neurosci 2009;63:257-65. doi: 10.1111/j.1440- 1819.2009.01945.x.
  72. Dean B. Understanding the role of inflammatory-related pathways in the pathophysiology and treatment of psychiatric disorders: Evidence from human peripheral studies and CNS studies. Int J Neuropsychopharmacol 2011;14:997-1012. doi: 10.1017/S1461145710001410.
  73. Meyer U. Anti-inflammatory signaling in schizophrenia. Brain Behav Immun 2011;25:1507-18. doi: 10.1016/j. bbi.2011.05.014.
  74. Monji A, Kato TA, Mizoguchi Y, Horikawa H, Seki Y, Kasai M, et al. Neuroinflammation in schizophrenia especially focused on the role of microglia. Prog Neuropsychopharmacol Biol Psychiatry 2013;42:115- 21. doi: 10.1016/j.pnpbp.2011.12.002.
  75. Keller WR, Kum LM, Wehring HJ, Koola MM, Buchanan RW, Kelly DL. A review of anti-inflammatory agents for symptoms of schizophrenia. J Psychopharmacol 2013;27:337-42. doi: 10.1177/0269881112467089.
  76. Levkovitz Y, Mendlovich S, Riwkes S, Braw Y, Levkovitch-Verbin H, Gal G, et al. A double-blind, randomized study of minocycline for the treatment of negative and cognitive symptoms in early-phase schizophrenia. J Clin Psychiatry 2010;71:138-49. doi: 10.4088/JCP.08m04666yel.
  77. Sofuoglu M, Mooney M, Kosten T, Waters A, Hashimoto K. Minocycline attenuates subjective rewarding effects of dextroamphetamine in humans. Psychopharmacology (Berl) 2011;213:61-8. doi: 10.1007/s00213-010-2014-5.
  78. Bagcioglu E, Solmaz V, Erbas O, Özkul B, Çakar B, Uyanikgil Y, et al. Modafinil improves autism-like behavior in rats by reducing neuroinflammation. J Neuroimmune Pharmacol 2023;18:9-23. doi: 10.1007/ s11481-023-10061-2.
  79. Lai MC, Lombardo MV, Baron-Cohen S. Autism. Lancet 2014;383:896-910. doi: 10.1016/S0140- 6736(13)61539-1.
  80. Baio J, Wiggins L, Christensen DL, Maenner MJ, Daniels J, Warren Z, et al. Prevalence of autism spectrum disorder among children aged 8 years - autism and developmental disabilities monitoring network, 11 sites, United States, 2014. MMWR Surveill Summ 2018;67:1-23. doi: 10.15585/mmwr.ss6706a1.
  81. Rodier PM. Environmental exposures that increase the risk of autism spectrum disorders. Autism Spectrum Disorders. Oxford: Oxford University Press; 2011.
  82. Elsabbagh M, Divan G, Koh YJ, Kim YS, Kauchali S, Marcín C, et al. Global prevalence of autism and other pervasive developmental disorders. Autism Res 2012;5:160-79. doi: 10.1002/aur.239.
  83. Werling DM, Geschwind DH. Sex differences in autism spectrum disorders. Curr Opin Neurol 2013;26:146-53. doi: 10.1097/WCO.0b013e32835ee548.
  84. Mandell DS, Novak MM, Zubritsky CD. Factors associated with age of diagnosis among children with autism spectrum disorders. Pediatrics 2005;116:1480-6. doi: 10.1542/peds.2005-0185.
  85. Volkmar FR, McPartland JC. From Kanner to DSM-5: Autism as an evolving diagnostic concept. Annu Rev Clin Psychol 2014;10:193-212. doi: 10.1146/annurevclinpsy-032813-153710.
  86. Johnson CP, Myers SM; American Academy of Pediatrics Council on Children With Disabilities. Identification and evaluation of children with autism spectrum disorders. Pediatrics 2007;120:1183-215. doi: 10.1542/ peds.2007-2361.
  87. Bilousova TV, Dansie L, Ngo M, Aye J, Charles JR, Ethell DW, et al. Minocycline promotes dendritic spine maturation and improves behavioural performance in the fragile X mouse model. J Med Genet 2009;46:94- 102. doi: 10.1136/jmg.2008.061796.
  88. Siller SS, Broadie K. Neural circuit architecture defects in a Drosophila model of Fragile X syndrome are alleviated by minocycline treatment and genetic removal of matrix metalloproteinase. Dis Model Mech 2011;4:673-85. doi: 10.1242/dmm.008045.
  89. Tassone F, Iong KP, Tong TH, Lo J, Gane LW, BerryKravis E, et al. FMR1 CGG allele size and prevalence ascertained through newborn screening in the United States. Genome Med 2012;4:100. doi: 10.1186/gm401.
  90. Jacobs PA, Glover TW, Mayer M, Fox P, Gerrard JW, Dunn HG, et al. X-linked mental retardation: A study of 7 families. Am J Med Genet 1980;7:471-89. doi: 10.1002/ajmg.1320070408.
  91. Paribello C, Tao L, Folino A, Berry-Kravis E, Tranfaglia M, Ethell IM, et al. Open-label add-on treatment trial of minocycline in fragile X syndrome. BMC Neurol 2010;10:91. doi: 10.1186/1471-2377-10-91.
  92. Leigh MJ, Nguyen DV, Mu Y, Winarni TI, Schneider A, Chechi T, et al. A randomized double-blind, placebocontrolled trial of minocycline in children and adolescents with fragile x syndrome. J Dev Behav Pediatr 2013;34:147- 55. doi: 10.1097/DBP.0b013e318287cd17.
  93. Kumar H, Sharma B. Minocycline ameliorates prenatal valproic acid induced autistic behaviour, biochemistry and blood brain barrier impairments in rats. Brain Res 2016;1630:83-97. doi: 10.1016/j.brainres.2015.10.052.
  94. Lovelace JW, Ethell IM, Binder DK, Razak KA. Minocycline treatment reverses sound evoked EEG abnormalities in a mouse model of fragile x syndrome. Front Neurosci 2020;14:771. doi: 10.3389/fnins.2020.00771.
  95. Pardo CA, Buckley A, Thurm A, Lee LC, Azhagiri A, Neville DM, et al. A pilot open-label trial of minocycline in patients with autism and regressive features. J Neurodev Disord 2013;5:9. doi: 10.1186/1866-1955-5-9.
  96. Ghaleiha A, Alikhani R, Kazemi MR, Mohammadi MR, Mohammadinejad P, Zeinoddini A, et al. Minocycline as adjunctive treatment to risperidone in children with autistic disorder: A randomized, double-blind placebocontrolled trial. J Child Adolesc Psychopharmacol 2016;26:784-91. doi: 10.1089/cap.2015.0175.
  97. Shigemori T, Sakai A, Takumi T, Itoh Y, Suzuki H. Altered microglia in the amygdala are involved in anxiety-related behaviors of a copy number variation mouse model of autism. J Nippon Med Sch 2015;82:92- 9. doi: 10.1272/jnms.82.92.