1
|
Wu Z, Yin Y, Liu R, Li X, Wang Z, Wu C, Tan J, Fu Z, Song C, Lee Wong N, Peng X, Lai S, Cui J, Han M, Peng Y, Sun Y, Wu L, Adzic M, Zeng L, Zhang H, Yau SY, Chen G. Chronic treatment of mixture of two iridoids proportional to prescriptional dose of Yueju improves hippocampal PACAP-related neuroinflammation and neuroplasticity signaling in the LPS-induced depression model. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119031. [PMID: 39522842 DOI: 10.1016/j.jep.2024.119031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 10/30/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Geniposide (GP) and shanzhiside methyl ester (SM) are the two important bioactive compounds in the classical traditional Chinese herbal medicine Yueju Pill, which is currently used as an over-the-counter (OTC) medicine in China. Yueju has been demonstrated with antidepressant-like effects with the prescriptional dose. As GP and SM both have antidepressant potential, the synergism of them could be crucial to the function of Yueju. OBJECTIVES The neuropeptide pituitary adenylyl cyclase-activating polypeptide (PACAP) has been implicated in the onset of antidepressant-like response. Here we investigated the synergism of the chronic treatment with GP and SM, at proportional doses to Yueju, on antidepressant-like effects, and underlying mechanism of PACAP-related signaling in a neuroinflammation-based depression model. MATERIALS AND METHODS Depression-related behaviors were tested in the lipopolysaccharide (LPS)-induced depression model. The molecular signaling of neuroinflammation and neuroplasticity was investigated using Western blot analysis, immunofluorescence and pharmacological inhibition of mTOR signaling. RESULTS Chronic treatment of GP and SM (GS) at the dose which is proportional to the prescriptional dose of Yueju synergistically elicited antidepressant-like effects. Chronic treatment of the GS or the conventional antidepressant fluoxetine (FLX) showed antidepressant-like effects in LPS-injected mice. In vitro analysis indicated the synergism of GS on PACAP expression. In the hippocampus of LPS-injected mice, both GS and FLX enhanced PACAP expression, downregulated the inflammatory signaling of Iba-1/NF-кB/IL-1β and NLRP3, and upregulated the neuroplasticity signaling of mTOR-BDNF/PSD95. Additionally, both treatments reduced microglia activation indicated by Iba-1 immunofluorescent staining. Rapamycin, an mTOR inhibitor, blunted the antidepressant-like effects and the upregulation of BDNF expression induced by chronic GS. CONCLUSION The antidepressant-like effects elicited by chronic fluoxetine or by synergistic doses of GS were involved in the upregulation of hippocampal PACAP levels, in association with ameliorated neuroinflammation and neuroplasticity signaling in LPS-injected mice. GS synergism may play a key part in the antidepressant-like effects of the prescriptional dose of Yueju.
Collapse
Affiliation(s)
- Zhangjie Wu
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, School of Chinese Medicine, Jinan University, Guangzhou, 510632, China; Zhuhai Institute of Jinan University, Zhuhai, 519070, China; Departments of Psychiatry & Clinical and Translational Institute of Psychiatric Disorders, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China; Guangdong-Hong Kong-Macau Joint Laboratory of Traditional Chinese Medicine on Brain-Peripheral omeostasis and Comprehensive Health, Jinan University, Guangzhou, 510632, China
| | - Ying Yin
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, School of Chinese Medicine, Jinan University, Guangzhou, 510632, China; Zhuhai Institute of Jinan University, Zhuhai, 519070, China; Departments of Psychiatry & Clinical and Translational Institute of Psychiatric Disorders, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China; Guangdong-Hong Kong-Macau Joint Laboratory of Traditional Chinese Medicine on Brain-Peripheral omeostasis and Comprehensive Health, Jinan University, Guangzhou, 510632, China
| | - Ruiyi Liu
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, School of Chinese Medicine, Jinan University, Guangzhou, 510632, China; Zhuhai Institute of Jinan University, Zhuhai, 519070, China; Departments of Psychiatry & Clinical and Translational Institute of Psychiatric Disorders, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China; Guangdong-Hong Kong-Macau Joint Laboratory of Traditional Chinese Medicine on Brain-Peripheral omeostasis and Comprehensive Health, Jinan University, Guangzhou, 510632, China
| | - Xianhui Li
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, School of Chinese Medicine, Jinan University, Guangzhou, 510632, China; Zhuhai Institute of Jinan University, Zhuhai, 519070, China; Departments of Psychiatry & Clinical and Translational Institute of Psychiatric Disorders, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China; Guangdong-Hong Kong-Macau Joint Laboratory of Traditional Chinese Medicine on Brain-Peripheral omeostasis and Comprehensive Health, Jinan University, Guangzhou, 510632, China
| | - Ziying Wang
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, School of Chinese Medicine, Jinan University, Guangzhou, 510632, China; Zhuhai Institute of Jinan University, Zhuhai, 519070, China; Departments of Psychiatry & Clinical and Translational Institute of Psychiatric Disorders, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China; Guangdong-Hong Kong-Macau Joint Laboratory of Traditional Chinese Medicine on Brain-Peripheral omeostasis and Comprehensive Health, Jinan University, Guangzhou, 510632, China
| | - Changyu Wu
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, School of Chinese Medicine, Jinan University, Guangzhou, 510632, China; Zhuhai Institute of Jinan University, Zhuhai, 519070, China; Departments of Psychiatry & Clinical and Translational Institute of Psychiatric Disorders, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China; Guangdong-Hong Kong-Macau Joint Laboratory of Traditional Chinese Medicine on Brain-Peripheral omeostasis and Comprehensive Health, Jinan University, Guangzhou, 510632, China
| | - Jingwen Tan
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, School of Chinese Medicine, Jinan University, Guangzhou, 510632, China; Zhuhai Institute of Jinan University, Zhuhai, 519070, China; Departments of Psychiatry & Clinical and Translational Institute of Psychiatric Disorders, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China; Guangdong-Hong Kong-Macau Joint Laboratory of Traditional Chinese Medicine on Brain-Peripheral omeostasis and Comprehensive Health, Jinan University, Guangzhou, 510632, China
| | - Zhenzhen Fu
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, School of Chinese Medicine, Jinan University, Guangzhou, 510632, China; Zhuhai Institute of Jinan University, Zhuhai, 519070, China; Departments of Psychiatry & Clinical and Translational Institute of Psychiatric Disorders, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China; Guangdong-Hong Kong-Macau Joint Laboratory of Traditional Chinese Medicine on Brain-Peripheral omeostasis and Comprehensive Health, Jinan University, Guangzhou, 510632, China
| | - Chenghao Song
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, School of Chinese Medicine, Jinan University, Guangzhou, 510632, China; Zhuhai Institute of Jinan University, Zhuhai, 519070, China; Departments of Psychiatry & Clinical and Translational Institute of Psychiatric Disorders, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China; Guangdong-Hong Kong-Macau Joint Laboratory of Traditional Chinese Medicine on Brain-Peripheral omeostasis and Comprehensive Health, Jinan University, Guangzhou, 510632, China
| | - Nga Lee Wong
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, School of Chinese Medicine, Jinan University, Guangzhou, 510632, China; Zhuhai Institute of Jinan University, Zhuhai, 519070, China; Departments of Psychiatry & Clinical and Translational Institute of Psychiatric Disorders, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China; Guangdong-Hong Kong-Macau Joint Laboratory of Traditional Chinese Medicine on Brain-Peripheral omeostasis and Comprehensive Health, Jinan University, Guangzhou, 510632, China
| | - Xiangyi Peng
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, School of Chinese Medicine, Jinan University, Guangzhou, 510632, China; Zhuhai Institute of Jinan University, Zhuhai, 519070, China; Departments of Psychiatry & Clinical and Translational Institute of Psychiatric Disorders, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China; Guangdong-Hong Kong-Macau Joint Laboratory of Traditional Chinese Medicine on Brain-Peripheral omeostasis and Comprehensive Health, Jinan University, Guangzhou, 510632, China
| | - Shixiong Lai
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, School of Chinese Medicine, Jinan University, Guangzhou, 510632, China; Zhuhai Institute of Jinan University, Zhuhai, 519070, China; Departments of Psychiatry & Clinical and Translational Institute of Psychiatric Disorders, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China; Guangdong-Hong Kong-Macau Joint Laboratory of Traditional Chinese Medicine on Brain-Peripheral omeostasis and Comprehensive Health, Jinan University, Guangzhou, 510632, China
| | - Jinshuai Cui
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, School of Chinese Medicine, Jinan University, Guangzhou, 510632, China; Zhuhai Institute of Jinan University, Zhuhai, 519070, China; Departments of Psychiatry & Clinical and Translational Institute of Psychiatric Disorders, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China; Guangdong-Hong Kong-Macau Joint Laboratory of Traditional Chinese Medicine on Brain-Peripheral omeostasis and Comprehensive Health, Jinan University, Guangzhou, 510632, China
| | - Mingzhi Han
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, School of Chinese Medicine, Jinan University, Guangzhou, 510632, China; Zhuhai Institute of Jinan University, Zhuhai, 519070, China; Departments of Psychiatry & Clinical and Translational Institute of Psychiatric Disorders, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China; Guangdong-Hong Kong-Macau Joint Laboratory of Traditional Chinese Medicine on Brain-Peripheral omeostasis and Comprehensive Health, Jinan University, Guangzhou, 510632, China
| | - Yuhan Peng
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, School of Chinese Medicine, Jinan University, Guangzhou, 510632, China; Zhuhai Institute of Jinan University, Zhuhai, 519070, China; Departments of Psychiatry & Clinical and Translational Institute of Psychiatric Disorders, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China; Guangdong-Hong Kong-Macau Joint Laboratory of Traditional Chinese Medicine on Brain-Peripheral omeostasis and Comprehensive Health, Jinan University, Guangzhou, 510632, China
| | - Yan Sun
- Key Laboratory of Integrative Biomedicine for Brain Diseases, School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Lei Wu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, China
| | - Miroslav Adzic
- "Vinča Institute" of Nuclear Sciences, Laboratory of Molecular Biology and Endocrinology 090, University of Belgrade, 11001, Belgrade, Serbia
| | - Li Zeng
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, 999078, China
| | - Hailou Zhang
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, School of Chinese Medicine, Jinan University, Guangzhou, 510632, China; Zhuhai Institute of Jinan University, Zhuhai, 519070, China; Departments of Psychiatry & Clinical and Translational Institute of Psychiatric Disorders, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China; Guangdong-Hong Kong-Macau Joint Laboratory of Traditional Chinese Medicine on Brain-Peripheral omeostasis and Comprehensive Health, Jinan University, Guangzhou, 510632, China.
| | - Suk-Yu Yau
- Department of Rehabilitation Sciences, Hong Kong Polytechnic University, Hong Kong, 999077, China; Mental Health Research Center (MHRC), The Hong Kong Polytechnic University, Hong Kong S.A.R, 999077, China.
| | - Gang Chen
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, School of Chinese Medicine, Jinan University, Guangzhou, 510632, China; Zhuhai Institute of Jinan University, Zhuhai, 519070, China; Departments of Psychiatry & Clinical and Translational Institute of Psychiatric Disorders, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China; Guangdong-Hong Kong-Macau Joint Laboratory of Traditional Chinese Medicine on Brain-Peripheral omeostasis and Comprehensive Health, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
2
|
Sherpa NN, De Giorgi R, Ostinelli EG, Choudhury A, Dolma T, Dorjee S. Efficacy and safety profile of oral creatine monohydrate in add-on to cognitive-behavioural therapy in depression: An 8-week pilot, double-blind, randomised, placebo-controlled feasibility and exploratory trial in an under-resourced area. Eur Neuropsychopharmacol 2025; 90:28-35. [PMID: 39488067 DOI: 10.1016/j.euroneuro.2024.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 10/06/2024] [Accepted: 10/14/2024] [Indexed: 11/04/2024]
Abstract
Pre-clinical and clinical evidence proposes that creatine monohydrate, an affordable nutraceutical, could be a useful adjunct to conventional antidepressant treatments. In this pilot feasibility and exploratory study, we investigate the 8-week effects of creatine in addition to cognitive-behavioural therapy (CBT) versus placebo plus CBT in depression. For the primary efficacy outcome of change in Patient Health Questionnaire-9 depression score at study endpoint, we used mixed-model repeated measures analysis of covariance. Logistic regressions were employed to assess acceptability (any-cause dropouts), tolerability (dropouts for adverse events), and safety (patients experiencing one or more adverse events). We calculated effect sizes adjusted for age, sex, and baseline depression score. One-hundred participants (50 females, mean age= 30.4 ± 7.4 years) with depression (mean PHQ-9 = 17.6 ± 6.3) were randomised to either creatine+CBT (N = 50) or placebo+CBT (N = 50). At 8 weeks, PHQ-9 scores were lower in both study arms, but significantly more so in participants taking creatine (mean difference= -5.12). Treatment discontinuations due to any cause and to adverse events, and proportion of participants with at least one adverse event were comparable between study arms. This hypothesis-generating trial suggests that creatine could be a useful and safe supplement to CBT for depression. Longer and larger clinical trials are warranted.
Collapse
Affiliation(s)
- Nima Norbu Sherpa
- Department of Radiography and Podiatry, Glasgow Caledonian University, Cowcaddens Rd, Glasgow G4 0BA, United Kingdom; Division of Mental Health, Universal Human Rights and Social Development Association, Non-Government Organisation, Uttarakhand, 248001, India
| | - Riccardo De Giorgi
- Department of Psychiatry, University of Oxford, Warneford Hospital, Warneford Lane, Oxford, OX3 7JX, United Kingdom; Oxford Health NHS Foundation Trust, Warneford Hospital, Warneford Lane, Oxford, OX3 7JX, United Kingdom.
| | - Edoardo Giuseppe Ostinelli
- Department of Psychiatry, University of Oxford, Warneford Hospital, Warneford Lane, Oxford, OX3 7JX, United Kingdom; Oxford Health NHS Foundation Trust, Warneford Hospital, Warneford Lane, Oxford, OX3 7JX, United Kingdom; Oxford Precision Psychiatry Lab, NIHR Oxford Health Biomedical Research Centre, Oxford, OX3 7JX, United Kingdom
| | - Amrita Choudhury
- Department of Psychology, St Xavier's University, Kolkata, Action Area III-B, New Town, West Bengal, 700160, India
| | - Tenzin Dolma
- Division of Mental Health, Universal Human Rights and Social Development Association, Non-Government Organisation, Uttarakhand, 248001, India
| | - Sangila Dorjee
- Department of Psychiatry, New S.T.N.M Multi Speciality Government Hospital, Sikkim, 737101, India
| |
Collapse
|
3
|
Wu Z, Yin Y, Liu R, Li X, Sun Y, Yau SY, Wu L, Liu Y, Adzic M, Zhang H, Chen G. A refined formula derived from Jiawei-Xiaoyao pill exerts rapid antidepressant-like effects in LPS-induced depression by reducing neuroinflammation and restoring neuroplasticity signaling. JOURNAL OF ETHNOPHARMACOLOGY 2024; 335:118647. [PMID: 39094756 DOI: 10.1016/j.jep.2024.118647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 07/21/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Jiawei-Xiaoyao Pill (JWX), a classic formula in traditional Chinese medicine, is derived from Xiaoyao Pill by adding significant amounts of Gardeniae Fructus (GF) and Moutan Cortex (MC). It is frequently used for the treatment of depression. JWX has been demonstrated to uniquely elicit rapid antidepressant-like effects within the prescribed dosage range. To date, GF has been shown to have rapid antidepressant-like effects, but a much higher dose is required than its proportion in JWX. It is assumed that the synergism of GF with a minimum number of other herbs in JWX serves as a refined formula that exerts these rapid antidepressant-like effects. Identification of a refined formula is important for prioritizing the herbs and ingredients to optimize the quality control of JWX. However, such a refined formula for JWX has not been identified yet. AIM OF THE STUDY Here we aimed to identify a refined formula derived from JWX for optimized rapid antidepressant-like effects. Since the neuroinflammation mechanism involving in depression treatment has not been previously investigated for JWX, we tested the mechanism for both JWX and the refined formula. MATERIALS AND METHODS Individual herbs (MC; ASR, Angelica Sinensis Radix; Bupleuri Radix; Paeonia Radix Alba) that show antidepressant-like responses were mixed with GF at the proportional dosage in JWX to identify the refined formula. Rapid antidepressant-like effects were assessed by using NSF (Novelty Suppressed Feeding Test) and other behavioral tests following a single administration. The identified formula was further tested in a lipopolysaccharide (LPS)-induced depressive model, and the molecular signaling mechanisms were investigated using Western blot analysis, immunofluorescence, and pharmacological inhibition of mTOR signaling. Scopolamine (Scop) was used as a positive control for induction of rapid antidepressant effects. RESULTS A combination of GF, MC and ASR (GMA) at their dosages proportional to JWX induced behavioral signs of rapid antidepressant-like responses in both normal and LPS-treated mice, with the antidepressant-like effects sustained for 5 d. Similar to JWX or Scop, GMA rapidly reduced the neuroinflammation signaling of Iba-1-NF-кB, enhanced neuroplasticity signaling of CaMKII-mTOR-BDNF, and attenuated the upregulated expressions of the NMDAR sub-units GluN1 and GluN2B in the hippocampus of LPS-treated mice. GMA, JWX and Scop rapidly restored the number of BDNF-positive cells reduced by LPS treatment in the CA3 region of the hippocampus. Furthermore, rapamycin, a selective inhibitor of mTOR, blunted the rapid antidepressant-like effects and hippocampal BDNF signaling upregulation by GMA. CONCLUSION GMA may serve as a refined formula from JWX, capable of inducing rapid antidepressant-like effects. In the LPS-induced depression model, the effects of GMA were mediated via rapidly alleviating neuroinflammation and enhancing neuroplasticity.
Collapse
Affiliation(s)
- Zhangjie Wu
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, School of Chinese Medicine, Jinan University, Guangzhou, 510632, PR China; Zhuhai Institute of Jinan University, Zhuhai, 519070, PR China; Guangdong-Hong Kong-Macao Joint Laboratory of Traditional Chinese Medicine on Brain-Peripheral Homeostasis and Comprehensive Health, Jinan University, Guangzhou, 510632, PR China; Departments of Psychiatry & Clinical and Translational Institute of Psychiatric Disorders, First Affiliated Hospital of Jinan University, Guangzhou, 510632, PR China
| | - Ying Yin
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, School of Chinese Medicine, Jinan University, Guangzhou, 510632, PR China; Zhuhai Institute of Jinan University, Zhuhai, 519070, PR China; Guangdong-Hong Kong-Macao Joint Laboratory of Traditional Chinese Medicine on Brain-Peripheral Homeostasis and Comprehensive Health, Jinan University, Guangzhou, 510632, PR China; Departments of Psychiatry & Clinical and Translational Institute of Psychiatric Disorders, First Affiliated Hospital of Jinan University, Guangzhou, 510632, PR China
| | - Ruiyi Liu
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, School of Chinese Medicine, Jinan University, Guangzhou, 510632, PR China; Zhuhai Institute of Jinan University, Zhuhai, 519070, PR China; Guangdong-Hong Kong-Macao Joint Laboratory of Traditional Chinese Medicine on Brain-Peripheral Homeostasis and Comprehensive Health, Jinan University, Guangzhou, 510632, PR China; Departments of Psychiatry & Clinical and Translational Institute of Psychiatric Disorders, First Affiliated Hospital of Jinan University, Guangzhou, 510632, PR China
| | - Xianhui Li
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, School of Chinese Medicine, Jinan University, Guangzhou, 510632, PR China; Zhuhai Institute of Jinan University, Zhuhai, 519070, PR China; Guangdong-Hong Kong-Macao Joint Laboratory of Traditional Chinese Medicine on Brain-Peripheral Homeostasis and Comprehensive Health, Jinan University, Guangzhou, 510632, PR China
| | - Yan Sun
- Key Laboratory of Integrative Biomedicine for Brain Diseases, School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Suk-Yu Yau
- Department of Rehabilitation Sciences, Hong Kong Polytechnic University, PR China
| | - Lei Wu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, PR China
| | - Yan Liu
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, School of Chinese Medicine, Jinan University, Guangzhou, 510632, PR China; Zhuhai Institute of Jinan University, Zhuhai, 519070, PR China
| | - Miroslav Adzic
- "Vinča Institute" of Nuclear Sciences, Laboratory of Molecular Biology and Endocrinology 090, University of Belgrade, 11001 Belgrade, Serbia
| | - Hailou Zhang
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, School of Chinese Medicine, Jinan University, Guangzhou, 510632, PR China; Zhuhai Institute of Jinan University, Zhuhai, 519070, PR China; Guangdong-Hong Kong-Macao Joint Laboratory of Traditional Chinese Medicine on Brain-Peripheral Homeostasis and Comprehensive Health, Jinan University, Guangzhou, 510632, PR China; Departments of Psychiatry & Clinical and Translational Institute of Psychiatric Disorders, First Affiliated Hospital of Jinan University, Guangzhou, 510632, PR China.
| | - Gang Chen
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, School of Chinese Medicine, Jinan University, Guangzhou, 510632, PR China; Zhuhai Institute of Jinan University, Zhuhai, 519070, PR China; Guangdong-Hong Kong-Macao Joint Laboratory of Traditional Chinese Medicine on Brain-Peripheral Homeostasis and Comprehensive Health, Jinan University, Guangzhou, 510632, PR China; Departments of Psychiatry & Clinical and Translational Institute of Psychiatric Disorders, First Affiliated Hospital of Jinan University, Guangzhou, 510632, PR China.
| |
Collapse
|
4
|
Zhang Y, Wei CK, Wang P, Zheng LC, Cheng Y, Ren ZH, Jin YH, Yao YY, Liu HZ. S-ketamine alleviates depression-like behavior and hippocampal neuroplasticity in the offspring of mice that experience prenatal stress. Sci Rep 2024; 14:26929. [PMID: 39505897 PMCID: PMC11542010 DOI: 10.1038/s41598-024-76226-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 10/11/2024] [Indexed: 11/08/2024] Open
Abstract
Prenatal stress exerts long-term impact on neurodevelopment in the offspring, with consequences such as increasing the offspring's risk of depression in adolescence and early adulthood. S-ketamine can produce rapid and robust antidepressant effects, but it is not clear yet whether and how S-ketamine alleviates depression in prenatally stressed offspring. The current study incestigated the preliminary anti-depression mechanism of S-ketamine in prenatally stressed offspring, particularly with regard to neuroplasticity. The pregnant females were given chronic unpredictable mild stress on the 7th-20th day of pregnancy and their male offspring were intraperitoneally injected with a single dose of S-ketamine (10 mg/kg) on postnatal day 42. Our findings showed that S-ketamine treatment counteracted the development of depression-like behaviors in prenatally stressed offspring. At the cellular level, S-ketamine markedly enhanced neuroplasticity in the CA1 hippocampus: Golgi-Cox staining showed that S-ketamine alleviated the reduction of neuronal complexity and dendritic spine density; Transmission electron microscopy indicated that S-ketamine reversed synaptic morphology alterations. At the molecular level, by western blot and RT-PCR we detected that S-ketamine significantly upregulated the expression of BDNF and PSD95 and activated AKT and mTOR in the hippocampus. In conclusion, prenatal stress induced by chronic unpredictable mild stress leads to depressive-like behaviors and hippocampal neuroplasticity impairments in male offspring. S-ketamine can produce antidepressant effects by enhancing hippocampal neuroplasticity via the BDNF/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Psychiatry, Changzhou Dean Hospital (also known as Changzhou No.9 People's Hospital), Changzhou, China
- Department of Psychiatry, Chaohu Hospital of Anhui Medical University, Hefei, China
| | - Chu-Ke Wei
- Department of Psychiatry, Anhui Psychiatric Center, Anhui Medical University, Hefei, China
| | - Ping Wang
- Department of Psychiatry, Chaohu Hospital of Anhui Medical University, Hefei, China
| | - Liu-Cheng Zheng
- Department of Psychiatry, Anhui Psychiatric Center, Anhui Medical University, Hefei, China
| | - Yang Cheng
- Department of Psychiatry, Anhui Psychiatric Center, Anhui Medical University, Hefei, China
| | - Zhen-Hua Ren
- Department of Anatomy, Anhui Medical University, Hefei, China
- Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei, China
| | - Yu-Hong Jin
- Department of Psychiatry, Changzhou Dean Hospital (also known as Changzhou No.9 People's Hospital), Changzhou, China
| | - Yu-You Yao
- School of Public Health, Anhui Medical University, Hefei, China.
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Hefei, China.
| | - Huan-Zhong Liu
- Department of Psychiatry, Chaohu Hospital of Anhui Medical University, Hefei, China.
- Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei, China.
| |
Collapse
|
5
|
Yang J, Yuan M, Zhang W. The major biogenic amine metabolites in mood disorders. Front Psychiatry 2024; 15:1460631. [PMID: 39381610 PMCID: PMC11458445 DOI: 10.3389/fpsyt.2024.1460631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 09/04/2024] [Indexed: 10/10/2024] Open
Abstract
Mood disorders, including major depressive disorder and bipolar disorder, have a profound impact on more than 300 million people worldwide. It has been demonstrated mood disorders were closely associated with deviations in biogenic amine metabolites, which are involved in numerous critical physiological processes. The peripheral and central alteration of biogenic amine metabolites in patients may be one of the potential pathogeneses of mood disorders. This review provides a concise overview of the latest research on biogenic amine metabolites in mood disorders, such as histamine, kynurenine, and creatine. Further studies need larger sample sizes and multi-center collaboration. Investigating the changes of biogenic amine metabolites in mood disorders can provide biological foundation for diagnosis, offer guidance for more potent treatments, and aid in elucidating the biological mechanisms underlying mood disorders.
Collapse
Affiliation(s)
- Jingyi Yang
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Minlan Yuan
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Zhang
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
- Big Data Center, Sichuan University, Chengdu, China
| |
Collapse
|
6
|
Talaee N, Azadvar S, Khodadadi S, Abbasi N, Asli-Pashaki ZN, Mirabzadeh Y, Kholghi G, Akhondzadeh S, Vaseghi S. Comparing the effect of fluoxetine, escitalopram, and sertraline, on the level of BDNF and depression in preclinical and clinical studies: a systematic review. Eur J Clin Pharmacol 2024; 80:983-1016. [PMID: 38558317 DOI: 10.1007/s00228-024-03680-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 03/21/2024] [Indexed: 04/04/2024]
Abstract
Brain-derived neurotrophic factor (BDNF) dysfunction is one of the most important mechanisms underlying depression. It seems that selective serotonin reuptake inhibitors (SSRIs) improve depression via affecting BDNF level. In this systematic review, for the first time, we aimed to review the effect of three SSRIs including fluoxetine, escitalopram, and sertraline, on both depression and BDNF level in preclinical and clinical studies. PubMed electronic database was searched, and 193 articles were included in this study. After reviewing all manuscripts, only one important difference was found: subjects. We found that SSRIs induce different effects in animals vs. humans. Preclinical studies showed many controversial effects, while human studies showed only two effects: improvement of depression, with or without the improvement of BDNF. However, most studies used chronic SSRIs treatment, while acute SSRIs were not effectively used and evaluated. In conclusion, it seems that SSRIs are reliable antidepressants, and the improvement effect of SSRIs on depression is not dependent to BDNF level (at least in human studies).
Collapse
Affiliation(s)
- Nastaran Talaee
- Department of Psychology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Shataw Azadvar
- Department of Power Electronic, Faculty of Electrical Engineering, Sahand University of Technology, Tabriz, Iran
| | - Sanaz Khodadadi
- Student Research Committee, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Nahal Abbasi
- Department of Health Psychology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | | | - Yasaman Mirabzadeh
- Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Gita Kholghi
- Department of Psychology, Faculty of Human Sciences, Tonekabon Branch, Islamic Azad University, Tonekabon, Iran
| | - Shahin Akhondzadeh
- Psychiatric Research Center, Department of Psychiatry, Faculty of Medicine, Roozbeh Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Salar Vaseghi
- Cognitive Neuroscience Lab, Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, 1419815477, Iran.
| |
Collapse
|
7
|
Chen F, Li L, Huang M, Wang Y, Wang L, Jin F, Yang L, Gao M, Li L, Wang Y, Zhou L, Yang J, Yao G, Li Q, Yang X. Natural product Kaji-ichigoside F1 exhibits rapid antidepression via activating the AMPA-BDNF-mTOR pathway and inhibiting the NMDAR-CaMKIIα pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 126:155452. [PMID: 38422650 DOI: 10.1016/j.phymed.2024.155452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 01/31/2024] [Accepted: 02/11/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND Depression is a common and recurrent neuropsychiatric disorder. Recent studies have shown that the N-methyl-d-aspartate (NMDA) receptor (NMDAR) is involved in the pathophysiology of depression. Previous studies have found that Kaji-ichigoside F1 (KF1) has a protective effect against NMDA-induced neurotoxicity. However, the antidepressant mechanism of KF1 has not been confirmed yet. PURPOSE In the present study, we aimed to evaluate the rapid antidepressant activity of KF1 and explore the underlying mechanism. STUDY DESIGN First, we explored the effect of KF1 on NMDA-induced hippocampal neurons and the underlying mechanism. Second, depression was induced in C57BL/6 mice via chronic unpredictable mild stress (CUMS), and the immediate and persistent depression-like behavior was evaluated using the forced swimming test (FST) after a single administration of KF1. Third, the contributions of NMDA signaling to the antidepressant effect of KF1 were investigated using pharmacological interventions. Fourth, CUMS mice were treated with KF1 for 21 days, and then their depression-like behaviors and the underlying mechanism were further explored. METHODS The FST was used to evaluate immediate and persistent depression-like behavior after a single administration of KF1 with or without NMDA pretreatment. The effect of KF1 on depressive-like behavior was investigated in CUMS mice by treating them with KF1 once daily for 21 days through the sucrose preference test, FST, open field test, and tail suspension test. Then, the effects of KF1 on the morphology and molecular and functional phenotypes of primary neuronal cells and hippocampus of mice were investigated by hematoxylin-eosin staining, Nissl staining, propidium iodide staining, TUNEL staining, Ca2+ imaging, JC-1 staining, ELISA, immunofluorescence analysis, RT-PCR, and Western blot. RESULTS KF1 could effectively improve cellular viability, reduce apoptosis, inhibit the release of LDH and Ca2+, and increase the mitochondrial membrane potential and the number of dendritic spines numbers in hippocampal neurons. Moreover, behavioral tests showed that KF1 exerted acute and sustained antidepressant-like effects by reducing Glu-levels and ameliorating neuronal damage in the hippocampus. Additionally, in vivo and in vitro experiments revealed that PSD95, Syn1, α-amino-3‑hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA), and brain-derived neurotrophic factor (BDNF) were upregulated at the protein level, and BDNF and AMPA were upregulated at the mRNA level. NR1 and NR2A showed the opposite trend. CONCLUSION These results confirm that KF1 exerts rapid antidepressant effects mainly by activating the AMPA-BDNF-mTOR pathway and inhibiting the NMDAR-CaMKIIα pathway. This study serves as a new reference for discovering rapid antidepressants.
Collapse
Affiliation(s)
- Faju Chen
- School of Basic Medical Sciences/State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; Engineering Research Center of Natural Product Efficient Utilization in Guizhou, Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Liangqun Li
- School of Basic Medical Sciences/State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; Engineering Research Center of Natural Product Efficient Utilization in Guizhou, Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Maoyang Huang
- Engineering Research Center of Natural Product Efficient Utilization in Guizhou, Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Yuankai Wang
- Huaxi District People,s Hospital, Guiyang 550025, China
| | - Li Wang
- School of Basic Medical Sciences/State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; Engineering Research Center of Natural Product Efficient Utilization in Guizhou, Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Fengli Jin
- Engineering Research Center of Natural Product Efficient Utilization in Guizhou, Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Lishou Yang
- School of Basic Medical Sciences/State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; Engineering Research Center of Natural Product Efficient Utilization in Guizhou, Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Ming Gao
- School of Basic Medical Sciences/State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; Engineering Research Center of Natural Product Efficient Utilization in Guizhou, Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Lilang Li
- School of Basic Medical Sciences/State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; Engineering Research Center of Natural Product Efficient Utilization in Guizhou, Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Yu Wang
- School of Basic Medical Sciences/State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; Engineering Research Center of Natural Product Efficient Utilization in Guizhou, Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Lang Zhou
- School of Basic Medical Sciences/State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; Engineering Research Center of Natural Product Efficient Utilization in Guizhou, Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Juan Yang
- School of Basic Medical Sciences/State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; Engineering Research Center of Natural Product Efficient Utilization in Guizhou, Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Guanping Yao
- School of Basic Medical Sciences/State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; Engineering Research Center of Natural Product Efficient Utilization in Guizhou, Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Qiji Li
- School of Basic Medical Sciences/State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; Engineering Research Center of Natural Product Efficient Utilization in Guizhou, Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Xiaosheng Yang
- School of Basic Medical Sciences/State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; Engineering Research Center of Natural Product Efficient Utilization in Guizhou, Natural Products Research Center of Guizhou Province, Guiyang 550014, China.
| |
Collapse
|
8
|
Zhang H, Sun Y, Huang Z, Wu Z, Ying Y, Liu R, Lin J, Li C, Chen G. Jiawei-Xiaoyao pill elicits a rapid antidepressant effect, dependent on activating CaMKII/mTOR/BDNF signaling pathway in the hippocampus. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:117016. [PMID: 37567427 DOI: 10.1016/j.jep.2023.117016] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 08/04/2023] [Accepted: 08/07/2023] [Indexed: 08/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Jiawei-Xiaoyao pill (JWX), a traditional Chinese medicine, was recorded in ancient Chinese medicine pharmacopoeia using for treatment of various diseases, including mood disorders. Current mainstream antidepressants have a disadvantage in delayed onset of action. The rapid antidepressant potential of JWX and the underlying mechanisms remain unclear. AIM OF THE STUDY We aimed to assess the rapid antidepressant potential of JWX, within the prescription dose range, and the distinct underlying neuroplasticity signaling mechanism. MATERIALS AND METHODS The rapid antidepressant response of JWX were determined using various behavioral paradigms, and in a corticosterone (CORT)-induced depression model in mice. The molecular neuroplasticity signaling and the expression of BDNF in the hippocampus was evaluated using immunoblotting and immunostaining. The contribution of specific signaling was investigated using pharmacological interventions. RESULTS A single dose of JWX induced rapid and persistent antidepressant effects in both the normal and chronic CORT-exposed mice. The phosphorylation of CaMKII, mTOR, ERK and the expressions of BDNF, synapsin1 and PSD95 increased at 30 min post JWX. JWX restored the expression of BDNF in the hippocampal dentate gyrus reduced by CORT-exposure. The rapid antidepressant effect and upregulation of BDNF expression by JWX was blunted by a mTOR antagonist, rapamycin, or a CaMKII antagonist, KN-93. CaMKII signaling blockade blunted mTOR signaling activated by JWX, but not vice versa. CONCLUSION JWX elicits a rapid antidepressant effect, via quickly stimulating CaMKII signaling, subsequently activating mTOR-BDNF signaling pathway, and thus enhancing hippocampal neuroplasticity.
Collapse
Affiliation(s)
- Hailou Zhang
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, Jinan University, Guangzhou, 510632, China; Departments of Psychiatry & Clinical and Translational Institute of Psychiatric Disorders, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Yan Sun
- Key Laboratory of Integrative Biomedicine for Brain Diseases, School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zihao Huang
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, Jinan University, Guangzhou, 510632, China; Departments of Psychiatry & Clinical and Translational Institute of Psychiatric Disorders, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Zhangjie Wu
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, Jinan University, Guangzhou, 510632, China; Departments of Psychiatry & Clinical and Translational Institute of Psychiatric Disorders, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Yin Ying
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, Jinan University, Guangzhou, 510632, China; Departments of Psychiatry & Clinical and Translational Institute of Psychiatric Disorders, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Ruiyi Liu
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, Jinan University, Guangzhou, 510632, China; Departments of Psychiatry & Clinical and Translational Institute of Psychiatric Disorders, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Juan Lin
- Guangzhou Pharmaceutical Holdings Limited., Guangzhou Baiyun Mountain and Hutchison Whampoa Ltd., Guangzhou, 510515, China
| | - Chuyuan Li
- Guangzhou Pharmaceutical Holdings Limited., Guangzhou Baiyun Mountain and Hutchison Whampoa Ltd., Guangzhou, 510515, China.
| | - Gang Chen
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, Jinan University, Guangzhou, 510632, China; Departments of Psychiatry & Clinical and Translational Institute of Psychiatric Disorders, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
9
|
Zhornitsky S, Oliva HNP, Jayne LA, Allsop ASA, Kaye AP, Potenza MN, Angarita GA. Changes in synaptic markers after administration of ketamine or psychedelics: a systematic scoping review. Front Psychiatry 2023; 14:1197890. [PMID: 37435405 PMCID: PMC10331617 DOI: 10.3389/fpsyt.2023.1197890] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 06/05/2023] [Indexed: 07/13/2023] Open
Abstract
Background Ketamine and psychedelics have abuse liability. They can also induce "transformative experiences" where individuals experience enhanced states of awareness. This enhanced awareness can lead to changes in preexisting behavioral patterns which could be beneficial in the treatment of substance use disorders (SUDs). Preclinical and clinical studies suggest that ketamine and psychedelics may alter markers associated with synaptic density, and that these changes may underlie effects such as sensitization, conditioned place preference, drug self-administration, and verbal memory performance. In this scoping review, we examined studies that measured synaptic markers in animals and humans after exposure to ketamine and/or psychedelics. Methods A systematic search was conducted following PRISMA guidelines, through PubMed, EBSCO, Scopus, and Web of Science, based on a published protocol (Open Science Framework, DOI: 10.17605/OSF.IO/43FQ9). Both in vivo and in vitro studies were included. Studies on the following synaptic markers were included: dendritic structural changes, PSD-95, synapsin-1, synaptophysin-1, synaptotagmin-1, and SV2A. Results Eighty-four studies were included in the final analyses. Seventy-one studies examined synaptic markers following ketamine treatment, nine examined psychedelics, and four examined both. Psychedelics included psilocybin/psilocin, lysergic acid diethylamide, N,N-dimethyltryptamine, 2,5-dimethoxy-4-iodoamphetamine, and ibogaine/noribogaine. Mixed findings regarding synaptic changes in the hippocampus and prefrontal cortex (PFC) have been reported when ketamine was administered in a single dose under basal conditions. Similar mixed findings were seen under basal conditions in studies that used repeated administration of ketamine. However, studies that examined animals during stressful conditions found that a single dose of ketamine counteracted stress-related reductions in synaptic markers in the hippocampus and PFC. Repeated administration of ketamine also counteracted stress effects in the hippocampus. Psychedelics generally increased synaptic markers, but results were more consistently positive for certain agents. Conclusion Ketamine and psychedelics can increase synaptic markers under certain conditions. Heterogeneous findings may relate to methodological differences, agents administered (or different formulations of the same agent), sex, and type of markers. Future studies could address seemingly mixed results by using meta-analytical approaches or study designs that more fully consider individual differences.
Collapse
Affiliation(s)
- Simon Zhornitsky
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
- Clinical Neuroscience Research Unit, Connecticut Mental Health Center, New Haven, CT, United States
| | - Henrique N. P. Oliva
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
- Clinical Neuroscience Research Unit, Connecticut Mental Health Center, New Haven, CT, United States
| | - Laura A. Jayne
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
- Clinical Neuroscience Research Unit, Connecticut Mental Health Center, New Haven, CT, United States
| | - Aza S. A. Allsop
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
- Clinical Neuroscience Research Unit, Connecticut Mental Health Center, New Haven, CT, United States
| | - Alfred P. Kaye
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
- Connecticut Mental Health Center, New Haven, CT, United States
- Clinical Neurosciences Division, VA National Center for PTSD, West Haven, CT, United States
| | - Marc N. Potenza
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
- Connecticut Mental Health Center, New Haven, CT, United States
- Child Study Center, Yale University School of Medicine, New Haven, CT, United States
- Department of Neuroscience, Yale University, New Haven, CT, United States
- Connecticut Council on Problem Gambling, Hartford, CT, United States
- Wu Tsai Institute, Yale University, New Haven, CT, United States
| | - Gustavo A. Angarita
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
- Clinical Neuroscience Research Unit, Connecticut Mental Health Center, New Haven, CT, United States
| |
Collapse
|
10
|
Camargo A, Dalmagro AP, Altê GA, Zeni ALB, Tasca CI, Rodrigues ALS. NMDA receptor-mediated modulation on glutamine synthetase and glial glutamate transporter GLT-1 is involved in the antidepressant-like and neuroprotective effects of guanosine. Chem Biol Interact 2023; 375:110440. [PMID: 36878458 DOI: 10.1016/j.cbi.2023.110440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 02/14/2023] [Accepted: 03/03/2023] [Indexed: 03/07/2023]
Abstract
Guanosine has been reported to elicit antidepressant-like responses in rodents, but if these actions are associated with its ability to afford neuroprotection against glutamate-induced toxicity still needs to be fully understood. Therefore, this study investigated the antidepressant-like and neuroprotective effects elicited by guanosine in mice and evaluated the possible involvement of NMDA receptors, glutamine synthetase, and GLT-1 in these responses. We found that guanosine (0.05 mg/kg, but not 0.01 mg/kg, p. o.) was effective in producing an antidepressant-like effect and protecting hippocampal and prefrontocortical slices against glutamate-induced damage. Our results also unveiled that ketamine (1 mg/kg, but not 0.1 mg/kg, i. p, an NMDA receptor antagonist) effectively elicited antidepressant-like actions and protected hippocampal and prefrontocortical slices against glutamatergic toxicity. Furthermore, the combined administration of sub-effective doses of guanosine (0.01 mg/kg, p. o.) with ketamine (0.1 mg/kg, i. p.) promoted an antidepressant-like effect and augmented glutamine synthetase activity and GLT-1 immunocontent in the hippocampus, but not in the prefrontal cortex. Our results also showed that the combination of sub-effective doses of ketamine and guanosine, at the same protocol schedule that exhibited an antidepressant-like effect, effectively abolished glutamate-induced damage in hippocampal and prefrontocortical slices. Our in vitro results reinforce that guanosine, ketamine, or sub-effective concentrations of guanosine plus ketamine protect against glutamate exposure by modulating glutamine synthetase activity and GLT-1 levels. Finally, molecular docking analysis suggests that guanosine might interact with NMDA receptors at the ketamine or glycine/d-serine co-agonist binding sites. These findings provide support for the premise that guanosine has antidepressant-like effects and should be further investigated for depression management.
Collapse
Affiliation(s)
- Anderson Camargo
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, 88040-900, Santa Catarina, Brazil
| | - Ana P Dalmagro
- Department of Natural Sciences, Center of Natural and Exact Sciences, Universidade Regional de Blumenau, Blumenau CEP, 89030-903, Santa Catarina, Brazil
| | - Glorister A Altê
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, 88040-900, Santa Catarina, Brazil
| | - Ana Lúcia B Zeni
- Department of Natural Sciences, Center of Natural and Exact Sciences, Universidade Regional de Blumenau, Blumenau CEP, 89030-903, Santa Catarina, Brazil
| | - Carla I Tasca
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, 88040-900, Santa Catarina, Brazil
| | - Ana Lúcia S Rodrigues
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, 88040-900, Santa Catarina, Brazil.
| |
Collapse
|
11
|
Suárez Santiago JE, Roldán GR, Picazo O. Ketamine as a pharmacological tool for the preclinical study of memory deficit in schizophrenia. Behav Pharmacol 2023; 34:80-91. [PMID: 36094064 DOI: 10.1097/fbp.0000000000000689] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Schizophrenia is a serious neuropsychiatric disorder characterized by the presence of positive symptoms (hallucinations, delusions, and disorganization of thought and language), negative symptoms (abulia, alogia, and affective flattening), and cognitive impairment (attention deficit, impaired declarative memory, and deficits in social cognition). Dopaminergic hyperactivity seems to explain the positive symptoms, but it does not completely clarify the appearance of negative and cognitive clinical manifestations. Preclinical data have demonstrated that acute and subchronic treatment with NMDA receptor antagonists such as ketamine (KET) represents a useful model that resembles the schizophrenia symptomatology, including cognitive impairment. This latter has been explained as a hypofunction of NMDA receptors located on the GABA parvalbumin-positive interneurons (near to the cortical pyramidal cells), thus generating an imbalance between the inhibitory and excitatory activity in the corticomesolimbic circuits. The use of behavioral models to explore alterations in different domains of memory is vital to learn more about the neurobiological changes that underlie schizophrenia. Thus, to better understand the neurophysiological mechanisms involved in cognitive impairment related to schizophrenia, the purpose of this review is to analyze the most recent findings regarding the effect of KET administration on these processes.
Collapse
Affiliation(s)
- José Eduardo Suárez Santiago
- Escuela Superior de Medicina, Laboratorio de Farmacología Conductual, Instituto Politécnico Nacional
- Facultad de Medicina, Departamento de Fisiología, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Gabriel Roldán Roldán
- Facultad de Medicina, Departamento de Fisiología, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Ofir Picazo
- Escuela Superior de Medicina, Laboratorio de Farmacología Conductual, Instituto Politécnico Nacional
| |
Collapse
|
12
|
Guanosine as a promising target for fast-acting antidepressant responses. Pharmacol Biochem Behav 2022; 218:173422. [PMID: 35732211 DOI: 10.1016/j.pbb.2022.173422] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/24/2022] [Accepted: 06/16/2022] [Indexed: 02/07/2023]
Abstract
Although the rapid-onset and sustained antidepressant responses elicited by ketamine have gained considerable attention in recent years, it has some knock-on effects that limit its widespread clinical use. Therefore, ketamine is considered the prototype for the new generation of glutamate-based rapid-acting antidepressants. Within this context, it has been demonstrated that guanosine, an endogenous guanine-based purine, has overlapping mechanisms of action with ketamine and is effective in eliciting fast antidepressant-like responses and even potentiating ketamine's actions in preclinical studies. Here, we review the recent findings regarding the ability of guanosine to produce rapid-acting antidepressant-like effects and we provide an overview of the molecular mechanisms underlying its antidepressant-like actions. Moreover, the neurobiological mechanisms underpinning the ability of guanosine in boosting the antidepressant-like and pro-synaptogenic effects elicited by ketamine are also reported. Taken together, this review opens perspectives for the use of guanosine alone or in combination with ketamine for the management of treatment-resistant depression.
Collapse
|
13
|
Depressive-like behaviors induced by chronic cerebral hypoperfusion associate with a dynamic change of GABA B1/B2 receptors expression in hippocampal CA1 region. Physiol Behav 2022; 254:113887. [PMID: 35724927 DOI: 10.1016/j.physbeh.2022.113887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/07/2022] [Accepted: 06/16/2022] [Indexed: 11/21/2022]
Abstract
Cerebral ischemia could induce depressive-like behaviors; however, the alteration of gamma-aminobutyric acid receptors type B (GABAB) receptors in these pathological processes has not been extensively investigated. The aim of the current study was to document the behavioral change and the alteration of GABAB receptors in chronic cerebral hypoperfusion. The permanent occlusion of the bilateral common carotid arteries (two-vessel occlusion, 2VO) was performed to induce chronic cerebral ischemia (CCH). The depressive-like behaviors were evaluated with sucrose preference test, novelty suppress feeding test as well as forced swim test at 4, 8, and 12 weeks after the 2VO surgery. The total, surface and intracellular expressions of GABAB subunit 1 (GABAB1) and subunit 2 (GABAB2) in hippocampal CA1 were quantified by western blot. The depressive-like behaviors were observed in rats suffered from 4, 8, and 12 weeks 2VO in sucrose preference test, novelty suppress feeding test and forced swim test. In addition, the surface and total expression of GABAB1 in CA1 was reduced at 4 weeks after 2VO rather than 8 or 12 weeks. While the surface and total expression of GABAB2 in CA1 was decreased throughout the ischemia timeline (4, 8, and 12 weeks). Taken together, our findings suggested the potential roles of GABAB1 and GABAB2 subunits involved in depressive-like behaviors caused by chronic cerebral hypoperfusion.
Collapse
|
14
|
Shin J, Lee J, Choi J, Ahn BT, Jang SC, You SW, Koh DY, Maeng S, Cha SY. Rapid-Onset Antidepressant-Like Effect of Nelumbinis semen in Social Hierarchy Stress Model of Depression. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:6897359. [PMID: 35677378 PMCID: PMC9168086 DOI: 10.1155/2022/6897359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 04/11/2022] [Accepted: 04/20/2022] [Indexed: 11/17/2022]
Abstract
Depression is a disease with increasing prevalence worldwide, and it is necessary to develop a therapeutic agent with better efficacy than existing antidepressant drugs. Antidepressants that act on the glutamatergic nervous system, such as ketamine, have a rapid-onset antidepressant effect and are effective against treatment-resistant depression. However, because of the addictive potential of ketamine, alternative substances without psychological side effects are recommended. In particular, many natural compounds have been tested for their antidepressant effects. The antidepressant effects of Nelumbinis semen (NS) have been tested in many studies, along with the various actions of NS on the glutamatergic system. Thus, it was expected that NS might have a rapid-onset antidepressant effect. To test the antidepressant potential, despair and anhedonic behaviors were measured after administering NS to mice exposed to social hierarchy stress (SHS), and biochemical changes in the prefrontal cortex and hippocampus were analyzed. NS reduced despair-like responses in the forced swim test and tail suspension test. Mice exposed to SHS showed depression-like responses such as increased despair, reduced hedonia, and an anxiety-like response in the novelty suppressed feeding test. NS, but not fluoxetine, improved those depression-like behaviors after acute treatment, and NBQX, an AMPA receptor blocker, inhibited the antidepressant-like effects of NS. The antidepressant-like effect of NS was related to enhanced phosphorylation of mTOR in the prefrontal cortex and dephosphorylation of GluR1 S845 in the hippocampus. Since NS has shown antidepressant-like potential in a preclinical model, it may be considered as a candidate for the development of antidepressants in the future.
Collapse
Affiliation(s)
- Jihwan Shin
- Graduate School of East-West Medical Science, Kyung Hee University, Yong-in 17104, Republic of Korea
| | - Jeonghun Lee
- Graduate School of East-West Medical Science, Kyung Hee University, Yong-in 17104, Republic of Korea
| | - Junhyuk Choi
- Graduate School of East-West Medical Science, Kyung Hee University, Yong-in 17104, Republic of Korea
| | - Byung-Taek Ahn
- Graduate School of East-West Medical Science, Kyung Hee University, Yong-in 17104, Republic of Korea
| | - Sang Chul Jang
- Graduate School of East-West Medical Science, Kyung Hee University, Yong-in 17104, Republic of Korea
| | - Seung-Won You
- Graduate School of East-West Medical Science, Kyung Hee University, Yong-in 17104, Republic of Korea
| | - Do-Yeon Koh
- Graduate School of East-West Medical Science, Kyung Hee University, Yong-in 17104, Republic of Korea
| | - Sungho Maeng
- Graduate School of East-West Medical Science, Kyung Hee University, Yong-in 17104, Republic of Korea
- AgeTech-Service Convergence Major, Graduated School of East-West Medical Science, Kyung Hee University, Young-in 17104, Republic of Korea
| | - Seung-Yun Cha
- Graduate School of East-West Medical Science, Kyung Hee University, Yong-in 17104, Republic of Korea
| |
Collapse
|
15
|
Camargo A, Torrá ACNC, Dalmagro AP, Valverde AP, Kouba BR, Fraga DB, Alves EC, Rodrigues ALS. Prophylactic efficacy of ketamine, but not the low-trapping NMDA receptor antagonist AZD6765, against stress-induced maladaptive behavior and 4E-BP1-related synaptic protein synthesis impairment. Prog Neuropsychopharmacol Biol Psychiatry 2022; 115:110509. [PMID: 35033626 DOI: 10.1016/j.pnpbp.2022.110509] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/27/2021] [Accepted: 01/10/2022] [Indexed: 01/04/2023]
Abstract
Ketamine enhances the resilience against stress-induced depressive-like behavior, but its prophylactic efficacy in anxiety-related behaviors remains to be elucidated. Moreover, there is a need for developing novel preventive strategies against depressive- and anxiety-like behavior. AZD6765, a low-trapping NMDA receptor antagonist, shares with ketamine common molecular targets and produces rapid-onset antidepressant effects, suggesting that it could be a prophylactic agent. Therefore, this study investigated the prophylactic effect of ketamine against the depressive- and anxiety-like behavior induced by chronic restraint stress (2 h/day, for 10 days) in mice. We also investigated if AZD6765 exerts a resilience-enhancing response against these maladaptive behaviors. The contribution of 4E-BP1-related synaptic proteins synthesis (PSD-95/GluA1) in the possible pro-resilience efficacy of ketamine and AZD6765 was investigated. A single administration of ketamine (5 mg/kg, i.p.), but not AZD6765 (1 or 5 mg/kg, i.p.), given 1 week before the stress protocol, was effective in preventing stress-induced depressive-like behavior in the tail suspension test and splash test. Ketamine administered at 1 and 5 mg/kg (i.p.), but not AZD6765 (1 or 5 mg/kg, i.p.), prevented stress-induced anxiety-related self-grooming alterations. Stress-induced reduction on 4E-BP1 phosphorylation and PSD-95 and GluA1 immunocontent in the prefrontal cortex was prevented by ketamine (5 mg/kg, i.p.), but not AZD6765 (1 or 5 mg/kg, i.p.). The results indicate that ketamine, but not AZD6765, exerts a pro-resilience response against stress-induced maladaptive behavior, reinforcing that it could be a prophylactic agent to manage individuals at-risk to develop MDD and anxiety.
Collapse
Affiliation(s)
- Anderson Camargo
- Department of Biochemistry, Center of Biological Sciences, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Ana Clara N C Torrá
- Department of Biochemistry, Center of Biological Sciences, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Ana Paula Dalmagro
- Department of Natural Sciences, Center of Natural and Exact Sciences, Department of Natural Sciences, Regional University of Blumenau, Blumenau, SC, Brazil
| | - Ana Paula Valverde
- Department of Biochemistry, Center of Biological Sciences, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Bruna R Kouba
- Department of Biochemistry, Center of Biological Sciences, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Daiane B Fraga
- Department of Biochemistry, Center of Biological Sciences, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Eloise C Alves
- Department of Biochemistry, Center of Biological Sciences, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Ana Lúcia S Rodrigues
- Department of Biochemistry, Center of Biological Sciences, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, SC, Brazil.
| |
Collapse
|
16
|
Camargo A, Dalmagro AP, Delanogare E, Fraga DB, Wolin IAV, Zeni ALB, Brocardo PS, Rodrigues ALS. Guanosine boosts the fast, but not sustained, antidepressant-like and pro-synaptogenic effects of ketamine by stimulating mTORC1-driven signaling pathway. Eur Neuropsychopharmacol 2022; 57:15-29. [PMID: 35008015 DOI: 10.1016/j.euroneuro.2021.12.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 12/23/2021] [Accepted: 12/27/2021] [Indexed: 12/23/2022]
Abstract
The mTORC1-dependent dendritic spines formation represents a key mechanism for fast and long-lasting antidepressant responses, but it remains to be determined whether this mechanism may account for the ability of guanosine in potentiating ketamine's actions. Here, we investigated the ability of ketamine plus guanosine to elicit fast and sustained antidepressant-like and pro-synaptogenic effects in mice and the role of mTORC1 signaling in these responses. The combined administration of subthreshold doses of ketamine (0.1 mg/kg, i.p.) and guanosine (0.01 mg/kg, p.o.) caused a fast (1 h - 24 h), but not long-lasting (7 days) reduction in the immobility time in the tail suspension test. This behavioral effect was paralleled by a rapid (started in 1 h) and transient (back to baseline in 24 h) increase on BDNF, p-Akt (Ser473), p-GSK-3β (Ser9), p-mTORC1 (Ser2448), p-p70S6K (Thr389) immunocontent in the hippocampus, but not in the prefrontal cortex. Conversely, ketamine plus guanosine increased PSD-95 and GluA1 immunocontent in the prefrontal cortex, but not the hippocampus after 1 h, whereas increased levels of these proteins in both brain structures were observed after 24 h, but these effects did not persist after 7 days. The combined administration of ketamine plus guanosine raised the dendritic spines density in the ventral hippocampal DG and prefrontal cortex after 24 h Rapamycin (0.2 nmol/site, i.c.v.) abrogated the antidepressant-like effect and pro-synaptogenic responses triggered by ketamine plus guanosine. These results indicate that guanosine may boost the antidepressant-like effect of ketamine for up to 24 h by a mTORC1-dependent mechanism.
Collapse
Affiliation(s)
- Anderson Camargo
- Neuroscience Graduate Program, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, 88040-900 SC, Brazil
| | - Ana Paula Dalmagro
- Laboratory of Evaluation of Bioactive Substances, Department of Natural Sciences, Universidade Regional de Blumenau, 89030-903 Blumenau, SC, Brazil
| | - Eslen Delanogare
- Neuroscience Graduate Program, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, 88040-900 SC, Brazil
| | - Daiane B Fraga
- Neuroscience Graduate Program, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, 88040-900 SC, Brazil
| | - Ingrid A V Wolin
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, 88040-900 SC, Brazil
| | - Ana Lúcia B Zeni
- Laboratory of Evaluation of Bioactive Substances, Department of Natural Sciences, Universidade Regional de Blumenau, 89030-903 Blumenau, SC, Brazil
| | - Patricia S Brocardo
- Neuroscience Graduate Program, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, 88040-900 SC, Brazil; Department of Morphological Sciences, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, 88040-900 SC, Brazil
| | - Ana Lúcia S Rodrigues
- Neuroscience Graduate Program, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, 88040-900 SC, Brazil; Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, 88040-900 SC, Brazil.
| |
Collapse
|
17
|
Effects of Creatine Supplementation on Brain Function and Health. Nutrients 2022; 14:nu14050921. [PMID: 35267907 PMCID: PMC8912287 DOI: 10.3390/nu14050921] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 02/06/2023] Open
Abstract
While the vast majority of research involving creatine supplementation has focused on skeletal muscle, there is a small body of accumulating research that has focused on creatine and the brain. Preliminary studies indicate that creatine supplementation (and guanidinoacetic acid; GAA) has the ability to increase brain creatine content in humans. Furthermore, creatine has shown some promise for attenuating symptoms of concussion, mild traumatic brain injury and depression but its effect on neurodegenerative diseases appears to be lacking. The purpose of this narrative review is to summarize the current body of research pertaining to creatine supplementation on total creatine and phophorylcreatine (PCr) content, explore GAA as an alternative or adjunct to creatine supplementation on brain creatine uptake, assess the impact of creatine on cognition with a focus on sleep deprivation, discuss the effects of creatine supplementation on a variety of neurological and mental health conditions, and outline recent advances on creatine supplementation as a neuroprotective supplement following traumatic brain injury or concussion.
Collapse
|
18
|
Tao X, Wu S, Tang W, Li L, Huang L, Mo D, Liu C, Song T, Wang S, Wang J, He J. Alleviative effects of foraging exercise on depressive-like behaviors in chronic mild stress-induced ischemic rat model. Brain Inj 2022; 36:127-136. [PMID: 35138197 DOI: 10.1080/02699052.2022.2034949] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Poststroke depression (PSD) is a common complication that seriously affects the functional recovery and prognosis of an individual. As some patients with PSD fail to respond to drug therapy, it is urgent to find a viable alternative treatment. METHODS An active exercise program known as foraging exercise (FE), using food as bait, was designed. First, focal ischemia and chronic unpredictable mild stress (CUMS) were used to establish a PSD model in rats. FE was then performed for 4 weeks. Body weight and behavioral assessments were conducted at the end of the 4th and 8th weeks. RESULTS After 8 weeks, the results revealed that, compared with the PSD group, the behavioral scores of the rats in the PSD/FE group were significantly improved, the expression of Iba-1 in the affected frontal lobe and striatum was decreased, and serum levels of IL-6 and the IL-6/IL-10 ratio were downregulated. However, the ratio of residual brain volume in rats that had experienced CUMS was significantly less than that in the stroke group. CONCLUSION FE can alleviate the behavioral scores of PSD rats, and its mechanism may be related to a modulation of the immune-inflammation response of microglia. Furthermore, chronic, persistent stress may increase the volume of cerebral infarction after stroke.
Collapse
Affiliation(s)
- Xi Tao
- Department of Neurological Rehabilitation, Hunan Provincial People's Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, China
| | - Siyuan Wu
- Department of Neurological Rehabilitation, Hunan Provincial People's Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, China
| | - Wenjing Tang
- Department of Neurological Rehabilitation, Hunan Provincial People's Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, China
| | - Lu Li
- Department of Neurological Rehabilitation, Hunan Provincial People's Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, China
| | - Lijun Huang
- Department of Neurological Rehabilitation, Hunan Provincial People's Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, China
| | - Danheng Mo
- Department of Neurology, Hunan Provincial People's Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, China
| | - Chujuan Liu
- Department of Neurological Rehabilitation, Hunan Provincial People's Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, China
| | - Tao Song
- Department of Neurological Rehabilitation, Hunan Provincial People's Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, China
| | - Shuling Wang
- Hunan Provincial Institute of Geriatrics, Hunan Provincial People's Hospital, Hunan Normal University, Changsha, Hunan Province, China
| | - Jia Wang
- Department of Scientific Research, Hunan Provincial People's Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, China
| | - Juan He
- Department of Neurosurgery, Hunan Provincial People's Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, China
| |
Collapse
|
19
|
Kang MJY, Hawken E, Vazquez GH. The Mechanisms Behind Rapid Antidepressant Effects of Ketamine: A Systematic Review With a Focus on Molecular Neuroplasticity. Front Psychiatry 2022; 13:860882. [PMID: 35546951 PMCID: PMC9082546 DOI: 10.3389/fpsyt.2022.860882] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 03/18/2022] [Indexed: 12/25/2022] Open
Abstract
The mechanism of action underlying ketamine's rapid antidepressant effects in patients with depression, both suffering from major depressive disorder (MDD) and bipolar disorder (BD), including treatment resistant depression (TRD), remains unclear. Of the many speculated routes that ketamine may act through, restoring deficits in neuroplasticity may be the most parsimonious mechanism in both human patients and preclinical models of depression. Here, we conducted a literature search using PubMed for any reports of ketamine inducing neuroplasticity relevant to depression, to identify cellular and molecular events, relevant to neuroplasticity, immediately observed with rapid mood improvements in humans or antidepressant-like effects in animals. After screening reports using our inclusion/exclusion criteria, 139 publications with data from cell cultures, animal models, and patients with BD or MDD were included (registered on PROSPERO, ID: CRD42019123346). We found accumulating evidence to support that ketamine induces an increase in molecules involved in modulating neuroplasticity, and that these changes are paired with rapid antidepressant effects. Molecules or complexes of high interest include glutamate, AMPA receptors (AMPAR), mTOR, BDNF/TrkB, VGF, eEF2K, p70S6K, GSK-3, IGF2, Erk, and microRNAs. In summary, these studies suggest a robust relationship between improvements in mood, and ketamine-induced increases in molecular neuroplasticity, particularly regarding intracellular signaling molecules.
Collapse
Affiliation(s)
- Melody J Y Kang
- Center of Neuroscience Studies (CNS), Queen's University, Kingston, ON, Canada
| | - Emily Hawken
- Department of Psychiatry, Queen's University School of Medicine, Kingston, ON, Canada.,Providence Care Hospital, Kingston, ON, Canada
| | - Gustavo Hector Vazquez
- Center of Neuroscience Studies (CNS), Queen's University, Kingston, ON, Canada.,Department of Psychiatry, Queen's University School of Medicine, Kingston, ON, Canada.,Providence Care Hospital, Kingston, ON, Canada
| |
Collapse
|
20
|
Camargo A, Dalmagro AP, Wolin IAV, Siteneski A, Zeni ALB, Rodrigues ALS. A low-dose combination of ketamine and guanosine counteracts corticosterone-induced depressive-like behavior and hippocampal synaptic impairments via mTORC1 signaling. Prog Neuropsychopharmacol Biol Psychiatry 2021; 111:110371. [PMID: 34089815 DOI: 10.1016/j.pnpbp.2021.110371] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 05/27/2021] [Accepted: 05/30/2021] [Indexed: 01/01/2023]
Abstract
Ketamine exhibits rapid and sustained antidepressant responses, but its repeated use may cause adverse effects. Augmentation strategies have been postulated to be useful for the management/reduction of ketamine's dose and its adverse effects. Based on the studies that have suggested that ketamine and guanosine may share overlapping mechanisms of action, the present study investigated the antidepressant-like effect of subthreshold doses of ketamine and guanosine in mice subjected to repeated administration of corticosterone (CORT) and the role of mTORC1 signaling for this effect. The ability of the treatment with ketamine (0.1 mg/kg, i.p.) plus guanosine (0.01 mg/kg, p.o.) to counteract the depressive-like behavior induced by CORT (20 mg/kg, p.o., for 21 days) in mice, was paralleled with the prevention of the CORT-induced reduction on BDNF levels, Akt (Ser473) and GSK-3β (Ser9) phosphorylation, and PSD-95, GluA1, and synapsin immunocontent in the hippocampus. No changes on mTORC1 and p70S6K immunocontent were found in the hippocampus and prefrontal cortex of any experimental group. No alterations on BDNF, Akt/GSK-3β, mTORC1/p70S6K, and synaptic proteins were observed in the prefrontal cortex of mice. The antidepressant-like and pro-synaptogenic effects elicited by ketamine plus guanosine were abolished by the pretreatment with rapamycin (0.2 nmol/site, i.c.v., a selective mTORC1 inhibitor). Our results showed that the combined administration of ketamine and guanosine at low doses counteracted CORT-induced depressive-like behavior and synaptogenic disturbances by activating mTORC1 signaling. This study supports the notion that the combined administration of guanosine and ketamine may be a useful therapeutic strategy for the management of MDD.
Collapse
Affiliation(s)
- Anderson Camargo
- Neuroscience Postgraduate Program, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, 88040-900, SC, Brazil; Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, 88040-900, SC, Brazil
| | - Ana Paula Dalmagro
- Laboratory of Evaluation of Bioactive Substances, Department of Natural Sciences, Universidade Regional de Blumenau, 89030-903, Blumenau, SC, Brazil
| | - Ingrid A V Wolin
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, 88040-900, SC, Brazil
| | - Aline Siteneski
- Neuroscience Postgraduate Program, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, 88040-900, SC, Brazil; Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, 88040-900, SC, Brazil
| | - Ana Lúcia B Zeni
- Laboratory of Evaluation of Bioactive Substances, Department of Natural Sciences, Universidade Regional de Blumenau, 89030-903, Blumenau, SC, Brazil
| | - Ana Lúcia S Rodrigues
- Neuroscience Postgraduate Program, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, 88040-900, SC, Brazil; Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, 88040-900, SC, Brazil.
| |
Collapse
|
21
|
Camargo A, Dalmagro AP, Wolin IAV, Kaster MP, Rodrigues ALS. The resilient phenotype elicited by ketamine against inflammatory stressors-induced depressive-like behavior is associated with NLRP3-driven signaling pathway. J Psychiatr Res 2021; 144:118-128. [PMID: 34619490 DOI: 10.1016/j.jpsychires.2021.09.057] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/21/2021] [Accepted: 09/29/2021] [Indexed: 01/29/2023]
Abstract
Ketamine has emerged as a prophylactic agent against depressive-like behavior induced by stress. However, the possible pro-resilience effects of ketamine against inflammatory stressors-induced depressive-like behavior and the signaling pathways associated with this response remain to be determined. Therefore, this study investigated the ability of prophylactic ketamine administration to produce a pro-resilience effect against the depressive-like behavior induced by lipopolysaccharide (LPS - 0.83 mg/kg, i.p.) and tumor necrosis factor-alpha (TNF-α - 0.1 fg/site, i.c.v.) administration in mice. The possible contribution of the NLRP3 inflammasome-driven signaling pathway to this effect was evaluated in the ventral hippocampus. A single administration of ketamine (5 mg/kg, i.p.) given 1 week before the LPS or TNF-α administration prevented the depressive-like behavior induced by these inflammatory stressors in the tail suspension test (TST) and splash test (SPT). On the other hand, a lower dose of ketamine (1 mg/kg, i.p.) failed to produce a similar effect. The administration of LPS, but not TNF-α, increased the immunocontent of the microglial marker Iba-1 in the ventral hippocampus. LPS increased the immunocontent of all proteins related to NLRP3 signaling, namely ASC, NLRP3, TXNIP, cleaved caspase-1, and IL-1β in this brain region, while TNF-α only increased ASC and NLRP3 immunocontent. Ketamine administered at the dose of 5 mg/kg, but not at 1 mg/kg, prevented the increase on the immunocontent of NLRP3 inflammasome complex components and regulators induced by LPS or TNF-α administration. Collectively, these findings suggest that ketamine elicits a pro-resilient phenotype against inflammatory stressors-induced depressive-like behavior, an effect associated with the suppression of the NLRP3 inflammasome-driven signaling pathway.
Collapse
Affiliation(s)
- Anderson Camargo
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, 88040-900, SC, Brazil.
| | - Ana Paula Dalmagro
- Department of Natural Sciences, Center of Exact and Natural Sciences, Universidade Regional de Blumenau, 89030-903, Blumenau, SC, Brazil.
| | - Ingrid A V Wolin
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, 88040-900, SC, Brazil.
| | - Manuella P Kaster
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, 88040-900, SC, Brazil.
| | - Ana Lúcia S Rodrigues
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, 88040-900, SC, Brazil.
| |
Collapse
|
22
|
Valverde AP, Camargo A, Rodrigues ALS. Agmatine as a novel candidate for rapid-onset antidepressant response. World J Psychiatry 2021; 11:981-996. [PMID: 34888168 PMCID: PMC8613765 DOI: 10.5498/wjp.v11.i11.981] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 06/09/2021] [Accepted: 08/23/2021] [Indexed: 02/06/2023] Open
Abstract
Major depressive disorder (MDD) is a disabling and highly prevalent mood disorder as well as a common cause of suicide. Chronic stress, inflammation, and intestinal dysbiosis have all been shown to play crucial roles in the pathophysiology of MDD. Although conventional antidepressants are widely used in the clinic, they can take weeks to months to produce therapeutic effects. The discovery that ketamine promotes fast and sustaining antidepressant responses is one of the most important breakthroughs in the pharmacotherapy of MDD. However, the adverse psychomimetic/dissociative and neurotoxic effects of ketamine discourage its chronic use. Therefore, agmatine, an endogenous glutamatergic modulator, has been postulated to elicit fast behavioral and synaptogenic effects by stimulating the mechanistic target of rapamycin complex 1 signaling pathway, similar to ketamine. However, recent evidence has demonstrated that the modulation of the NLR family pyrin domain containing 3 inflammasome and gut microbiota, which have been shown to play a crucial role in the pathophysiology of MDD, may also participate in the antidepressant-like effects of both ketamine and agmatine. This review seeks to provide evidence about the mechanisms that may underlie the fast antidepressant-like responses of agmatine in preclinical studies. Considering the anti-inflammatory properties of agmatine, it may also be further investigated as a useful compound for the management of MDD associated with a pro-inflammatory state. Moreover, the fast antidepressant-like response of agmatine noted in animal models should be investigated in clinical studies.
Collapse
Affiliation(s)
- Ana Paula Valverde
- Department of Biochemistry, Campus Universitário, Center for Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, SC 88040900, Brazil
| | - Anderson Camargo
- Department of Biochemistry, Campus Universitário, Center for Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, SC 88040900, Brazil
| | - Ana Lúcia S Rodrigues
- Department of Biochemistry, Campus Universitário, Center for Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, SC 88040900, Brazil
| |
Collapse
|
23
|
Moretti M, Rodrigues ALS. Functional role of ascorbic acid in the central nervous system: a focus on neurogenic and synaptogenic processes. Nutr Neurosci 2021; 25:2431-2441. [PMID: 34493165 DOI: 10.1080/1028415x.2021.1956848] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Ascorbic acid, a water-soluble vitamin, is highly concentrated in the brain and participates in neuronal modulation and regulation of central nervous system (CNS) homeostasis. Ascorbic acid has emerged as a neuroprotective compound against neurotoxicants and neurodegenerative diseases, including Alzheimer's disease, multiple sclerosis and amyotrophic lateral sclerosis. Moreover, it improves behavioral and biochemical alterations in psychiatric disorders, including schizophrenia, anxiety, major depressive disorder, and bipolar disorder. Some recent studies have advanced the knowledge on the mechanisms associated with the preventive and therapeutic effects of ascorbic acid by showing that they are linked to improved neurogenesis and synaptic plasticity. This review shows that ascorbic acid has the potential to regulate positively stem cell generation and proliferation. Moreover, it improves neuronal differentiation of precursors cells, promotes adult hippocampal neurogenesis, and has synaptogenic effects that are possibly linked to its protective or therapeutic effects in the brain.
Collapse
Affiliation(s)
- Morgana Moretti
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Ana Lúcia S Rodrigues
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| |
Collapse
|
24
|
Creatine Supplementation Upregulates mTORC1 Signaling and Markers of Synaptic Plasticity in the Dentate Gyrus While Ameliorating LPS-Induced Cognitive Impairment in Female Rats. Nutrients 2021; 13:nu13082758. [PMID: 34444918 PMCID: PMC8398736 DOI: 10.3390/nu13082758] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/06/2021] [Accepted: 08/10/2021] [Indexed: 11/27/2022] Open
Abstract
Mild cognitive impairment (MCI) designates the boundary area between cognitive function in natural aging and dementia, and this is viewed as a therapeutic window to prevent the occurrence of dementia. The current study investigated the neurocognitive effects of oral creatine (Cr) supplementation in young female Wistar rats that received intracerebroventricular injections of lipopolysaccharide (LPS) to mimic MCI. Neuromolecular changes within the dentate gyrus were analyzed following behavioral testing. We also investigated both neurocognitive and neuromolecular changes following Cr supplementation in the absence of LPS in young female Wistar rats to further investigate mechanisms. Interestingly, based on trial 2 of Barnes maze test, Cr supplementation ameliorated spatial learning and memory deficit induced by LPS, shown by decreased latency time and errors to reach the escape box (p < 0.0001, n = 12). Cr supplementation also attenuated recognition memory deficit induced by LPS, shown by increased amount of time taken to explore the new object (p = 0.002, n = 12) during novel object recognition testing. Within the dentate gyrus, Cr supplementation in LPS injected rats upregulated mTORC1 signaling (p = 0.026 for mTOR phosphorylation, p = 0.002 for p70S6K phosphorylation, n = 8) as well as the synapsin (p = 0.008) and PSD-95 synaptic proteins (p = 0.015), in comparisons to LPS injected rats. However, Cr supplementation failed to further enhance spatial memory and recognition memory in the absence of LPS. In conclusion, Cr ameliorates LPS-induced cognitive impairment in a rodent MCI model. Mechanistically, these phenotypic effects may, in part, be mitigated via an upregulation of mTORC1 signaling, and an enhancement in synaptogenesis in the dentate gyrus. While preliminary, these findings may inform future research investigating neurocognitive effects of Cr for MCI patients.
Collapse
|
25
|
Fraga DB, Camargo A, Olescowicz G, Azevedo Padilha D, Mina F, Budni J, Brocardo PS, Rodrigues ALS. A single administration of ascorbic acid rapidly reverses depressive-like behavior and hippocampal synaptic dysfunction induced by corticosterone in mice. Chem Biol Interact 2021; 342:109476. [PMID: 33872575 DOI: 10.1016/j.cbi.2021.109476] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/31/2021] [Accepted: 04/12/2021] [Indexed: 12/31/2022]
Abstract
Ketamine is the prototype for glutamate-based fast-acting antidepressants. The establishment of ketamine-like drugs is still a challenge and ascorbic acid has emerged as a candidate. This study investigated the ascorbic acid's ability to induce a fast antidepressant-like response and to improve hippocampal synaptic markers in mice subjected to chronic corticosterone (CORT) administration. CORT was administered for 21 days, followed by a single administration of ascorbic acid (1 mg ∕Kg, p.o.), ketamine (1 mg ∕Kg, i.p.) or fluoxetine (10 mg ∕Kg, p.o.) in mice. Depressive-like behavior, hippocampal synaptic proteins immunocontent, dendrite spines density in the dentate gyrus (DG) were analyzed 24 h following treatments. The administration of ascorbic acid or ketamine, but not fluoxetine, counteracted CORT-induced depressive-like behavior in the tail suspension test (TST). CORT administration reduced PSD-95, GluA1, and synapsin (synaptic markers) immunocontent, and these alterations were reversed by ascorbic acid or ketamine, but only ketamine reversed the CORT-induced reduction on GluA1 immunocontent. In the ventral and dorsal DG, CORT decreased filopodia-, thin- and stubby-shaped spines, while ascorbic acid and ketamine abolished this alteration only in filopodia spines. Ascorbic acid and ketamine increased mushroom-shaped spines density in ventral and dorsal DG. Therefore, the results show that a single administration of ascorbic acid, in a way similar to ketamine, rapidly elicits an antidepressant-like response and reverses hippocampal synaptic deficits caused by CORT, an effect associated with increased levels of synaptic proteins and dendritic remodeling.
Collapse
Affiliation(s)
- Daiane B Fraga
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Anderson Camargo
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Gislaine Olescowicz
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Dayane Azevedo Padilha
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Francielle Mina
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Josiane Budni
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Patricia S Brocardo
- Department of Morphological Sciences, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Ana Lúcia S Rodrigues
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, SC, Brazil.
| |
Collapse
|
26
|
Cholewinski T, Pereira D, Moerland M, Jacobs GE. MTORC1 signaling as a biomarker in major depressive disorder and its pharmacological modulation by novel rapid-acting antidepressants. Ther Adv Psychopharmacol 2021; 11:20451253211036814. [PMID: 34733478 PMCID: PMC8558816 DOI: 10.1177/20451253211036814] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 07/16/2021] [Indexed: 12/15/2022] Open
Abstract
Major depressive disorder (MDD) is a multifactorial psychiatric disorder with obscure pathophysiology. A biomarker-based approach in combination with standardized interview-based instruments is needed to identify MDD subtypes and novel therapeutic targets. Recent findings support the impairment of the mammalian target of rapamycin complex 1 (mTORC1) in MDD. No well-established biomarkers of mTORC1 disease- and treatment-modulated activity are currently available for use in early phase antidepressant drug (AD) development. This review aims to summarize biomarkers of mTORC1 activity in MDD and to suggest how these could be implemented in future early clinical trials on mTORC1 modulating ADs. Therefore, a PubMed-based narrative literature review of the mTORC1 involvement in MDD was performed. We have summarized recent pre-clinical and clinical findings linking the MDD to the impaired activity of several key biomarkers related to mTORC1. Also, cases of restoration of these impairments by classical ADs and novel fast-acting investigational ADs are summarized. The presented biomarkers may be used to monitor pharmacological effects by novel rapid-acting mTORC1-targeting ADs. Based on findings in the peripheral blood mononuclear cells, we argue that those may serve as an ex vivo model for evaluation of mTORC1 activity and propose the use of the summarized biomarkers for this purpose. This could both facilitate the selection of a pharmacodynamically active dose and guide future early clinical efficacy studies in MDD. In conclusion, this review provides a blueprint for the rational development of rapid-acting mTORC1-targeting ADs.
Collapse
Affiliation(s)
| | - Diana Pereira
- Centre for Human Drug Research, Leiden, The Netherlands
| | | | - Gabriel E Jacobs
- Centre for Human Drug Research, Zernikedreef 8, 2333 CL Leiden, The Netherlands
| |
Collapse
|