1
|
Lee SH, Hofstede RP, Noriega de la Colina A, Gunton JH, Bernstock JD, Traverso G. Implantable systems for neurological chronotherapy. Adv Drug Deliv Rev 2025; 221:115574. [PMID: 40187646 DOI: 10.1016/j.addr.2025.115574] [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: 12/01/2024] [Revised: 02/26/2025] [Accepted: 03/24/2025] [Indexed: 04/07/2025]
Abstract
Implantable systems for neurological chronotherapy are poised to revolutionize the treatment of central nervous system diseases and disorders. These devices enable precise, time-controlled drug delivery aligned with the body's circadian rhythms, optimizing therapeutic outcomes. By bypassing the blood-brain barrier, they achieve high local drug concentrations while minimizing systemic side effects, offering significant advantages for conditions where traditional therapies often fall short. Platforms like SynchroMed II and CraniUS showcase this innovation, providing programmable delivery for conditions such as epilepsy and glioblastoma, with customizable profiles ranging from continuous infusion to timed bolus administration. Preclinical and clinical studies underscore the efficacy of aligning drug delivery with circadian rhythms, enhancing outcomes in chrono-chemotherapy and anti-epileptic treatments. Despite their promise, challenges remain, including the invasiveness of implantation within the brain, device longevity, synchronization complexities, and cost(s). Accordingly, this review explores the current state of implantable neurological systems that may be leveraged for chronotherapy, their applications, limitations, and potential to transform neurological disease/disorder management.
Collapse
Affiliation(s)
- Seung Ho Lee
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Roemer Pott Hofstede
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | - John H Gunton
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Joshua D Bernstock
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Giovanni Traverso
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
2
|
Feyrer A, Kerkel K, Mlcochova E, Langguth B, Schecklmann M. No sex difference in the antidepressive effect of transcranial magnetic stimulation (TMS): results from a retrospective analysis of a large real-world sample. World J Biol Psychiatry 2025; 26:170-178. [PMID: 40272912 DOI: 10.1080/15622975.2025.2488357] [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: 12/18/2024] [Revised: 03/29/2025] [Accepted: 03/31/2025] [Indexed: 04/26/2025]
Abstract
OBJECTIVES There is only limited knowledge about the impact of sex on the effectiveness of repetitive transcranial magnetic stimulation (rTMS) in depressive disorders. Here, we analysed a large real-world sample of depressive patients with respect to potential sex-specific effects of rTMS treatment. METHODS Data of 984 patients (539 females/445 males) were analysed. Patients received various antidepressant TMS protocols, most of them 10Hz, 20 Hz, Theta burst or accelerated protocols over the left dorsolateral prefrontal cortex. Changes in Hamilton Depression Scale (HAMD) and Major Depression Inventory (MDI) scores as well as response and remission rates were compared between female and male patients. RESULTS There were no significant differences in any outcome between female and male patients. Response rates according to the HAMD-21 scores were 34.3% for females and 30.1% for males, according to the MDI 33.1% and 33.5% respectively. In an additional explorative analysis there was a tendency towards better outcome for females for the 20 Hz protocol. CONCLUSIONS The antidepressive effectiveness of rTMS does not differ between men and women.
Collapse
Affiliation(s)
- Antonia Feyrer
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Katharina Kerkel
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Eva Mlcochova
- University Medical Center, University of Regensburg, Regensburg, Germany
| | - Berthold Langguth
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Martin Schecklmann
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| |
Collapse
|
3
|
Chen L, Chen P, Xie Y, Guo J, Chen R, Guo Y, Fang C. Twelve-hour ultradian rhythmic reprogramming of gene expression in the human ovary during aging. J Assist Reprod Genet 2025; 42:545-561. [PMID: 39849236 PMCID: PMC11871189 DOI: 10.1007/s10815-024-03339-8] [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: 09/16/2024] [Accepted: 11/21/2024] [Indexed: 01/25/2025] Open
Abstract
BACKGROUND The 12-h ultradian rhythm plays a crucial role in metabolic homeostasis, but its role in ovarian aging has not been explored. This study investigates age-related changes in 12-h rhythmic gene expression across various human tissues, with a particular focus on the ovary. METHODS We analyzed transcriptomic data from the GTEx project to examine 12-h ultradian rhythmic gene expression across multiple peripheral human tissues, exploring sex-specific patterns and age-related reprogramming of both 12-h and 24-h rhythmic gene expression. RESULTS Our findings revealed sex-dimorphic patterns in 12-h rhythmic gene expression, with females exhibiting stronger 12-h rhythms than males. Midlife (ages 40-49) was identified as a critical period for the reprogramming of both 12-h and 24-h rhythmic gene expression. The ovary was notable among other organs due to its high number of genes exhibiting 12-h rhythmic expression and a distinct pattern of rhythmic gene expression reprogramming during aging. This reprogramming involved two gene subsets: one subset adopted de novo 12-h rhythms, while another subset shifted from 24-h rhythms in younger individuals to dual 12-h and 24-h rhythms in middle-aged individuals. Both subsets were primarily associated with angiogenesis. CONCLUSIONS This study is the first to report age-related reprogramming of 12-h rhythms in human tissues, with a particular focus on the amplification of 12-h rhythms in angiogenesis-related genes in the aging ovary. These findings provide novel insights into the mechanisms structured format of the abstract text underlying ovarian aging and suggest potential therapeutic strategies targeting rhythmic gene expression in the ovary.
Collapse
Affiliation(s)
- Lina Chen
- Center of Reproductive Medicine, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China
- Guangdong Engineering Technology Research Center of Fertility Preservation, Guangzhou, 510655, China
| | - Peigen Chen
- Center of Reproductive Medicine, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China
- Guangdong Engineering Technology Research Center of Fertility Preservation, Guangzhou, 510655, China
| | - Yun Xie
- Center of Reproductive Medicine, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China
- Guangdong Engineering Technology Research Center of Fertility Preservation, Guangzhou, 510655, China
| | - Jiayi Guo
- Center of Reproductive Medicine, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China
- Guangdong Engineering Technology Research Center of Fertility Preservation, Guangzhou, 510655, China
| | - Rouzhu Chen
- Center of Reproductive Medicine, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China
- Guangdong Engineering Technology Research Center of Fertility Preservation, Guangzhou, 510655, China
| | - Yingchun Guo
- Center of Reproductive Medicine, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China.
- Guangdong Engineering Technology Research Center of Fertility Preservation, Guangzhou, 510655, China.
| | - Cong Fang
- Center of Reproductive Medicine, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China.
- Guangdong Engineering Technology Research Center of Fertility Preservation, Guangzhou, 510655, China.
| |
Collapse
|
4
|
Burns JN, Jenkins AK, Xue X, Petersen KA, Ketchesin KD, Perez MS, Vadnie CA, Scott MR, Seney ML, Tseng GC, McClung CA. Comparative transcriptomic rhythms in the mouse and human prefrontal cortex. Front Neurosci 2025; 18:1524615. [PMID: 39872996 PMCID: PMC11769989 DOI: 10.3389/fnins.2024.1524615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 12/23/2024] [Indexed: 01/30/2025] Open
Abstract
Introduction Alterations in multiple subregions of the human prefrontal cortex (PFC) have been heavily implicated in psychiatric diseases. Moreover, emerging evidence suggests that circadian rhythms in gene expression are present across the brain, including in the PFC, and that these rhythms are altered in disease. However, investigation into the potential circadian mechanisms underlying these diseases in animal models must contend with the fact that the human PFC is highly evolved and specialized relative to that of rodents. Methods Here, we use RNA sequencing to lay the groundwork for translational studies of molecular rhythms through a sex-specific, cross species comparison of transcriptomic rhythms between the mouse medial PFC (mPFC) and two subregions of the human PFC, the anterior cingulate cortex (ACC) and the dorsolateral PFC (DLPFC). Results We find that while circadian rhythm signaling is conserved across species and subregions, there is a phase shift in the expression of core clock genes between the mouse mPFC and human PFC subregions that differs by sex. Furthermore, we find that the identity of rhythmic transcripts is largely unique between the mouse mPFC and human PFC subregions, with the most overlap (20%, 236 transcripts) between the mouse mPFC and the human ACC in females. Nevertheless, we find that basic biological processes are enriched for rhythmic transcripts across species, with key differences between regions and sexes. Discussion Together, this work highlights both the evolutionary conservation of transcriptomic rhythms and the advancement of the human PFC, underscoring the importance of considering cross-species differences when using animal models.
Collapse
Affiliation(s)
- Jennifer N. Burns
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States
| | - Aaron K. Jenkins
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Xiangning Xue
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, United States
| | - Kaitlyn A. Petersen
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States
| | - Kyle D. Ketchesin
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States
| | - Megan S. Perez
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Chelsea A. Vadnie
- David O. Robbins Neuroscience Program, Department of Psychology, Ohio Wesleyan University, Delaware, OH, United States
| | - Madeline R. Scott
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States
| | - Marianne L. Seney
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States
| | - George C. Tseng
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, United States
| | - Colleen A. McClung
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
5
|
Burns JN, Jenkins AK, Xue X, Petersen KA, Ketchesin KD, Perez MS, Vadnie CA, Scott MR, Seney ML, Tseng GC, McClung CA. Comparative transcriptomic rhythms in the mouse and human prefrontal cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.26.615154. [PMID: 39386590 PMCID: PMC11463408 DOI: 10.1101/2024.09.26.615154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Alterations in multiple subregions of the human prefrontal cortex (PFC) have been heavily implicated in psychiatric diseases. Moreover, emerging evidence suggests that circadian rhythms in gene expression are present across the brain, including in the PFC, and that these rhythms are altered in disease. However, investigation into the potential circadian mechanisms underlying these diseases in animal models must contend with the fact that the human PFC is highly evolved and specialized relative to that of rodents. Here, we use RNA sequencing to lay the groundwork for translational studies of molecular rhythms through a sex-specific, cross species comparison of transcriptomic rhythms between the mouse medial PFC (mPFC) and two subregions of the human PFC, the anterior cingulate cortex (ACC) and the dorsolateral PFC (DLPFC). We find that while circadian rhythm signaling is conserved across species and subregions, there is a phase shift in the expression of core clock genes between the mouse mPFC and human PFC subregions that differs by sex. Furthermore, we find that the identity of rhythmic transcripts is largely unique between the mouse mPFC and human PFC subregions, with the most overlap (20%, 236 transcripts) between the mouse mPFC and the human ACC in females. Nevertheless, we find that basic biological processes are enriched for rhythmic transcripts across species, with key differences between regions and sexes. Together, this work highlights both the evolutionary conservation of transcriptomic rhythms and the advancement of the human PFC, underscoring the importance of considering cross-species differences when using animal models.
Collapse
Affiliation(s)
- Jennifer N. Burns
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15261
| | - Aaron K. Jenkins
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15261
| | - Xiangning Xue
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15261
| | - Kaitlyn A. Petersen
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15261
| | - Kyle D. Ketchesin
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15261
| | - Megan S. Perez
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261
| | - Chelsea A. Vadnie
- David O. Robbins Neuroscience Program, Department of Psychology, Ohio Wesleyan University, Delaware, OH 43015
| | - Madeline R. Scott
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15261
| | - Marianne L. Seney
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15261
| | - George C. Tseng
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15261
| | - Colleen A. McClung
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15261
| |
Collapse
|
6
|
Burns JN, Jenkins AK, Yin R, Zong W, Vadnie CA, DePoy LM, Petersen KA, Tsyglakova M, Scott MR, Tseng GC, Huang YH, McClung CA. Molecular and cellular rhythms in excitatory and inhibitory neurons in the mouse prefrontal cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.05.601880. [PMID: 39005410 PMCID: PMC11245095 DOI: 10.1101/2024.07.05.601880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Previous studies have shown that there are rhythms in gene expression in the mouse prefrontal cortex (PFC); however, the contribution of different cell types and potential variation by sex has not yet been determined. Of particular interest are excitatory pyramidal cells and inhibitory parvalbumin (PV) interneurons, as interactions between these cell types are essential for regulating the excitation/inhibition balance and controlling many of the cognitive functions regulated by the PFC. In this study, we identify cell-type specific rhythms in the translatome of PV and pyramidal cells in the mouse PFC and assess diurnal rhythms in PV cell electrophysiological properties. We find that while core molecular clock genes are conserved and synchronized between cell types, pyramidal cells have nearly twice as many rhythmic transcripts as PV cells (35% vs. 18%). Rhythmic transcripts in pyramidal cells also show a high degree of overlap between sexes, both in terms of which transcripts are rhythmic and in the biological processes associated with them. Conversely, in PV cells, rhythmic transcripts from males and females are largely distinct. Moreover, we find sex-specific effects of phase on action potential properties in PV cells that are eliminated by environmental circadian disruption. Together, this study demonstrates that rhythms in gene expression and electrophysiological properties in the mouse PFC vary by both cell type and sex. Moreover, the biological processes associated with these rhythmic transcripts may provide insight into the unique functions of rhythms in these cells, as well as their selective vulnerabilities to circadian disruption.
Collapse
Affiliation(s)
- Jennifer N. Burns
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15261
| | - Aaron K. Jenkins
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15261
| | - RuoFei Yin
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15261
| | - Wei Zong
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15261
| | - Chelsea A. Vadnie
- David O. Robbins Neuroscience Program, Department of Psychology, Ohio Wesleyan University, Delaware, OH 43015
| | - Lauren M. DePoy
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15261
| | - Kaitlyn A Petersen
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15261
| | - Mariya Tsyglakova
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15261
| | - Madeline R. Scott
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15261
| | - George C. Tseng
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15261
| | - Yanhua H. Huang
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15261
| | - Colleen A. McClung
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15261
| |
Collapse
|
7
|
Juffre A, Gumz ML. Recent advances in understanding the kidney circadian clock mechanism. Am J Physiol Renal Physiol 2024; 326:F382-F393. [PMID: 38174377 PMCID: PMC11207534 DOI: 10.1152/ajprenal.00214.2023] [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: 07/24/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 01/05/2024] Open
Abstract
Circadian rhythms are endogenous biological oscillations that regulate various physiological processes in organisms, including kidney function. The kidney plays a vital role in maintaining homeostasis by regulating water and electrolyte balance, blood pressure, and excretion of metabolic waste products, all of which display circadian rhythmicity. For this reason, studying the circadian regulation of the kidney is important, and the time of day is a biological and experimental variable that must be considered. Over the past decade, considerable progress has been made in understanding the molecular mechanisms underlying circadian regulation within the kidney. In this review, the current knowledge regarding circadian rhythms in the kidney is explored, focusing on the molecular clock machinery, circadian control of renal functions, and the impact of disrupted circadian rhythms on kidney health. In addition, parameters that should be considered and future directions are outlined in this review.
Collapse
Affiliation(s)
- Alexandria Juffre
- Department of Physiology and Aging, University of Florida, Gainesville, Florida, United States
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, Florida, United States
| | - Michelle L Gumz
- Department of Physiology and Aging, University of Florida, Gainesville, Florida, United States
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, Florida, United States
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida, United States
| |
Collapse
|
8
|
Naveed M, Chao OY, Hill JW, Yang YM, Huston JP, Cao R. Circadian neurogenetics and its implications in neurophysiology, behavior, and chronomedicine. Neurosci Biobehav Rev 2024; 157:105523. [PMID: 38142983 PMCID: PMC10872425 DOI: 10.1016/j.neubiorev.2023.105523] [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: 09/05/2023] [Revised: 12/13/2023] [Accepted: 12/20/2023] [Indexed: 12/26/2023]
Abstract
The circadian rhythm affects multiple physiological processes, and disruption of the circadian system can be involved in a range of disease-related pathways. The genetic underpinnings of the circadian rhythm have been well-studied in model organisms. Significant progress has been made in understanding how clock genes affect the physiological functions of the nervous system. In addition, circadian timing is becoming a key factor in improving drug efficacy and reducing drug toxicity. The circadian biology of the target cell determines how the organ responds to the drug at a specific time of day, thus regulating pharmacodynamics. The current review brings together recent advances that have begun to unravel the molecular mechanisms of how the circadian clock affects neurophysiological and behavioral processes associated with human brain diseases. We start with a brief description of how the ubiquitous circadian rhythms are regulated at the genetic, cellular, and neural circuit levels, based on knowledge derived from extensive research on model organisms. We then summarize the latest findings from genetic studies of human brain disorders, focusing on the role of human clock gene variants in these diseases. Lastly, we discuss the impact of common dietary factors and medications on human circadian rhythms and advocate for a broader application of the concept of chronomedicine.
Collapse
Affiliation(s)
- Muhammad Naveed
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA; Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH 43614, USA
| | - Owen Y Chao
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA
| | - Jennifer W Hill
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH 43614, USA
| | - Yi-Mei Yang
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA; Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Joseph P Huston
- Center for Behavioral Neuroscience, Institute of Experimental Psychology, Heinrich-Heine University, 40225 Düsseldorf, Germany
| | - Ruifeng Cao
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA; Department of Neurology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA.
| |
Collapse
|
9
|
Sica V, Deryagin O, Smith JG, Muñoz-Canoves P. Circadian transcriptome processing and analysis: a workflow for muscle stem cells. FEBS Open Bio 2023; 13:1228-1237. [PMID: 37394994 DOI: 10.1002/2211-5463.13629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/27/2023] [Accepted: 05/11/2023] [Indexed: 07/04/2023] Open
Abstract
Circadian rhythms coordinate biological processes with Earth's 24-h daily light/dark cycle. In the last years, efforts in the field of chronobiology have sought to understand the ways in which the circadian clock controls transcription across tissues and cells. This has been supported by the development of different bioinformatic approaches that allow the identification of 24-h oscillating transcripts. This workflow aims to describe how to isolate muscle stem cells for RNA sequencing analysis from a typical circadian experiment and introduces bioinformatic tools suitable for the analysis of circadian transcriptomes.
Collapse
Affiliation(s)
- Valentina Sica
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Oleg Deryagin
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Jacob G Smith
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Pura Muñoz-Canoves
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Altos labs Inc, San Diego, CA, USA
| |
Collapse
|
10
|
Shulman D, Dubnov S, Zorbaz T, Madrer N, Paldor I, Bennett DA, Seshadri S, Mufson EJ, Greenberg DS, Loewenstein Y, Soreq H. Sex-specific declines in cholinergic-targeting tRNA fragments in the nucleus accumbens in Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.08.527612. [PMID: 36798311 PMCID: PMC9934682 DOI: 10.1101/2023.02.08.527612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Introduction Females with Alzheimer's disease (AD) suffer accelerated dementia and loss of cholinergic neurons compared to males, but the underlying mechanisms are unknown. Seeking causal contributors to both these phenomena, we pursued changes in tRNA fragments (tRFs) targeting cholinergic transcripts (CholinotRFs). Methods We analyzed small RNA-sequencing data from the nucleus accumbens (NAc) brain region which is enriched in cholinergic neurons, compared to hypothalamic or cortical tissues from AD brains; and explored small RNA expression in neuronal cell lines undergoing cholinergic differentiation. Results NAc CholinotRFs of mitochondrial genome origin showed reduced levels that correlated with elevations in their predicted cholinergic-associated mRNA targets. Single cell RNA seq from AD temporal cortices showed altered sex-specific levels of cholinergic transcripts in diverse cell types; inversely, human-originated neuroblastoma cells under cholinergic differentiation presented sex-specific CholinotRF elevations. Discussion Our findings support CholinotRFs contributions to cholinergic regulation, predicting their involvement in AD sex-specific cholinergic loss and dementia.
Collapse
Affiliation(s)
- Dana Shulman
- The Edmond & Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
- The Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
- The Rachel and Selim Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Serafima Dubnov
- The Edmond & Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
- The Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Tamara Zorbaz
- The Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Nimrod Madrer
- The Edmond & Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
- The Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Iddo Paldor
- The Neurosurgery Department, Shaare Zedek Medical Center, Jerusalem 9103102, Israel
| | - David A. Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, 600 South Paulina, Suite 1028, Chicago, IL 60612, USA
| | - Sudha Seshadri
- UT Health Medical Arts & Research Center, San Antonio, TX 78229, USA
| | - Elliott J. Mufson
- Barrow Neurological Institute, St. Joseph’s Medical Center, Phoenix, AZ, 85013, USA
| | - David S. Greenberg
- The Edmond & Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
- The Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Yonatan Loewenstein
- The Edmond & Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
- The Department of Neurobiology, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
- The Department of Cognitive Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
- The Federmann Center for the Study of Rationality, Jerusalem 9190401, Israel
| | - Hermona Soreq
- The Edmond & Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
- The Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| |
Collapse
|
11
|
Jia JL, Alshamsan B, Ng TL. Temozolomide Chronotherapy in Glioma: A Systematic Review. Curr Oncol 2023; 30:1893-1902. [PMID: 36826108 PMCID: PMC9955138 DOI: 10.3390/curroncol30020147] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 01/27/2023] [Accepted: 01/30/2023] [Indexed: 02/08/2023] Open
Abstract
Outcomes for patients with high-grade glioma remain poor. Temozolomide (TMZ) is the only drug approved for first-line treatment of glioblastoma multiforme, the most aggressive form of glioma. Chronotherapy highlights the potential benefit of timed TMZ administration. This is based on pre-clinical studies of enhanced TMZ-induced glioma cytotoxicity dependent on circadian, oscillating expression of key genes involved in apoptosis, DNA damage repair, and cell-cycle mediated cell death. The current systematic review's primary aim was to evaluate the efficacy and toxicity of TMZ chronotherapy. A systemic review of literature following PRISMA guidelines looking at clinical outcomes on TMZ chronotherapy on gliomas was performed. The search in the English language included three databases (PubMed, EMBASE, and Cochrane) and five conferences from 1946 to April 2022. Two independent reviewers undertook screening, data extraction, and risk-of-bias assessment. A descriptive analysis was conducted due to limited data. Of the 269 articles screened, two unique studies were eligible and underwent abstraction for survival and toxicity findings. Both studies-one a retrospective cohort study (n = 166) and the other a prospective randomized feasibility study (n = 35)-were conducted by the same academic group and suggested a trend for improved overall survival, but possibly increased toxicity when TMZ was administered in the morning (vs. evening). There was limited evidence suggesting possible therapeutic value from administering TMZ in the morning, which may be consistent with the pre-clinical observations of the importance of the timing of TMZ administration in vitro. Larger, pragmatic, prospective randomized controlled trials are needed to ascertain the value of TMZ chronotherapy to provide optimized and equitable care for this population.
Collapse
Affiliation(s)
- Jason L. Jia
- Core Internal Medicine Residency Program, Department of Medicine, University of Ottawa, Ottawa, ON K1H 8L6, Canada
| | - Bader Alshamsan
- Department of Medicine, College of Medicine, Qassim University, Buraydah P.O. Box 6655, Saudi Arabia
- Division of Medical Oncology, Department of Medicine, The Ottawa Hospital Cancer Centre, Ottawa, ON K1H 8L6, Canada
| | - Terry L. Ng
- Division of Medical Oncology, Department of Medicine, The Ottawa Hospital Cancer Centre, Ottawa, ON K1H 8L6, Canada
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| |
Collapse
|
12
|
Valeri J, O’Donovan SM, Wang W, Sinclair D, Bollavarapu R, Gisabella B, Platt D, Stockmeier C, Pantazopoulos H. Altered expression of somatostatin signaling molecules and clock genes in the hippocampus of subjects with substance use disorder. Front Neurosci 2022; 16:903941. [PMID: 36161151 PMCID: PMC9489843 DOI: 10.3389/fnins.2022.903941] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 08/16/2022] [Indexed: 11/13/2022] Open
Abstract
Substance use disorders are a debilitating group of psychiatric disorders with a high degree of comorbidity with major depressive disorder. Sleep and circadian rhythm disturbances are commonly reported in people with substance use disorder and major depression and associated with increased risk of relapse. Hippocampal somatostatin signaling is involved in encoding and consolidation of contextual memories which contribute to relapse in substance use disorder. Somatostatin and clock genes also have been implicated in depression, suggesting that these molecules may represent key converging pathways involved in contextual memory processing in substance use and major depression. We used hippocampal tissue from a cohort of subjects with substance use disorder (n = 20), subjects with major depression (n = 20), subjects with comorbid substance use disorder and major depression (n = 24) and psychiatrically normal control subjects (n = 20) to test the hypothesis that expression of genes involved in somatostatin signaling and clock genes is altered in subjects with substance use disorder. We identified decreased expression of somatostatin in subjects with substance use disorder and in subjects with major depression. We also observed increased somatostatin receptor 2 expression in subjects with substance use disorder with alcohol in the blood at death and decreased expression in subjects with major depression. Expression of the clock genes Arntl, Nr1d1, Per2 and Cry2 was increased in subjects with substance use disorder. Arntl and Nr1d1 expression in comparison was decreased in subjects with major depression. We observed decreased expression of Gsk3β in subjects with substance use disorder. Subjects with comorbid substance use disorder and major depression displayed minimal changes across all outcome measures. Furthermore, we observed a significant increase in history of sleep disturbances in subjects with substance use disorder. Our findings represent the first evidence for altered somatostatin and clock gene expression in the hippocampus of subjects with substance use disorder and subjects with major depression. Altered expression of these molecules may impact memory consolidation and contribute to relapse risk.
Collapse
Affiliation(s)
- Jake Valeri
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, United States
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, United States
| | - Sinead M. O’Donovan
- Department of Neuroscience, University of Toledo Medical Center, Toledo, OH, United States
| | - Wei Wang
- Department of Medicine and Neurology, Brigham and Women’s Hospital, Boston, MA, United States
| | - David Sinclair
- Department of Neuroscience, University of Toledo Medical Center, Toledo, OH, United States
| | - Ratna Bollavarapu
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, United States
| | - Barbara Gisabella
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, United States
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, United States
| | - Donna Platt
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, United States
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, United States
| | - Craig Stockmeier
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, United States
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, United States
| | - Harry Pantazopoulos
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, United States
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, United States
- *Correspondence: Harry Pantazopoulos,
| |
Collapse
|
13
|
Damato AR, Katumba RGN, Luo J, Atluri H, Talcott GR, Govindan A, Slat EA, Weilbaecher KN, Tao Y, Huang J, Butt OH, Ansstas G, Johanns TM, Chheda MG, Herzog ED, Rubin JB, Campian JL. A randomized feasibility study evaluating temozolomide circadian medicine in patients with glioma. Neurooncol Pract 2022; 9:193-200. [PMID: 35601970 PMCID: PMC9113320 DOI: 10.1093/nop/npac003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Background Gliomas are the most common primary brain tumor in adults. Current treatments involve surgery, radiation, and temozolomide (TMZ) chemotherapy; however, prognosis remains poor and new approaches are required. Circadian medicine aims to maximize treatment efficacy and/or minimize toxicity by timed delivery of medications in accordance with the daily rhythms of the patient. We published a retrospective study showing greater anti-tumor efficacy for the morning, relative to the evening, administration of TMZ in patients with glioblastoma. We conducted this prospective randomized trial to determine the feasibility, and potential clinical impact, of TMZ chronotherapy in patients with gliomas (NCT02781792). Methods Adult patients with gliomas (WHO grade II-IV) were enrolled prior to initiation of monthly TMZ therapy and were randomized to receive TMZ either in the morning (AM) before 10 am or in the evening (PM) after 8 pm. Pill diaries were recorded to measure compliance and FACT-Br quality of life (QoL) surveys were completed throughout treatment. Study compliance, adverse events (AE), and overall survival were compared between the two arms. Results A total of 35 evaluable patients, including 21 with GBM, were analyzed (18 AM patients and 17 PM patients). Compliance data demonstrated the feasibility of timed TMZ dosing. There were no significant differences in AEs, QoL, or survival between the arms. Conclusions Chronotherapy with TMZ is feasible. A larger study is needed to validate the effect of chronotherapy on clinical efficacy.
Collapse
Affiliation(s)
- Anna R Damato
- Department of Biology, Washington University, St Louis, Missouri, USA
| | - Ruth G N Katumba
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri, USA
| | - Jingqin Luo
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
- Siteman Cancer Center Biostatistics Core, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Himachandana Atluri
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri, USA
| | - Grayson R Talcott
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri, USA
| | - Ashwin Govindan
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri, USA
- John T. Milliken Department of Medicine, Washington University School of Medicine, St Louis, Missouri, USA
| | - Emily A Slat
- Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri, USA
| | - Katherine N Weilbaecher
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri, USA
| | - Yu Tao
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
- Siteman Cancer Center Biostatistics Core, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jiayi Huang
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri, USA
| | - Omar H Butt
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri, USA
| | - George Ansstas
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri, USA
| | - Tanner M Johanns
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri, USA
| | - Milan G Chheda
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri, USA
| | - Erik D Herzog
- Department of Biology, Washington University, St Louis, Missouri, USA
- Department of Neuroscience, Washington University School of Medicine, St Louis, Missouri, USA
| | - Joshua B Rubin
- Department of Pediatrics, Washington University School of Medicine, St Louis, Missouri, USA
- Department of Neuroscience, Washington University School of Medicine, St Louis, Missouri, USA
| | - Jian L Campian
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri, USA
| |
Collapse
|
14
|
McCarthy MJ, Gottlieb JF, Gonzalez R, McClung CA, Alloy LB, Cain S, Dulcis D, Etain B, Frey BN, Garbazza C, Ketchesin KD, Landgraf D, Lee H, Marie‐Claire C, Nusslock R, Porcu A, Porter R, Ritter P, Scott J, Smith D, Swartz HA, Murray G. Neurobiological and behavioral mechanisms of circadian rhythm disruption in bipolar disorder: A critical multi-disciplinary literature review and agenda for future research from the ISBD task force on chronobiology. Bipolar Disord 2022; 24:232-263. [PMID: 34850507 PMCID: PMC9149148 DOI: 10.1111/bdi.13165] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
AIM Symptoms of bipolar disorder (BD) include changes in mood, activity, energy, sleep, and appetite. Since many of these processes are regulated by circadian function, circadian rhythm disturbance has been examined as a biological feature underlying BD. The International Society for Bipolar Disorders Chronobiology Task Force (CTF) was commissioned to review evidence for neurobiological and behavioral mechanisms pertinent to BD. METHOD Drawing upon expertise in animal models, biomarkers, physiology, and behavior, CTF analyzed the relevant cross-disciplinary literature to precisely frame the discussion around circadian rhythm disruption in BD, highlight key findings, and for the first time integrate findings across levels of analysis to develop an internally consistent, coherent theoretical framework. RESULTS Evidence from multiple sources implicates the circadian system in mood regulation, with corresponding associations with BD diagnoses and mood-related traits reported across genetic, cellular, physiological, and behavioral domains. However, circadian disruption does not appear to be specific to BD and is present across a variety of high-risk, prodromal, and syndromic psychiatric disorders. Substantial variability and ambiguity among the definitions, concepts and assumptions underlying the research have limited replication and the emergence of consensus findings. CONCLUSIONS Future research in circadian rhythms and its role in BD is warranted. Well-powered studies that carefully define associations between BD-related and chronobiologically-related constructs, and integrate across levels of analysis will be most illuminating.
Collapse
Affiliation(s)
- Michael J. McCarthy
- UC San Diego Department of Psychiatry & Center for Circadian BiologyLa JollaCaliforniaUSA
- VA San Diego Healthcare SystemSan DiegoCaliforniaUSA
| | - John F. Gottlieb
- Department of PsychiatryFeinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Robert Gonzalez
- Department of Psychiatry and Behavioral HealthPennsylvania State UniversityHersheyPennsylvaniaUSA
| | - Colleen A. McClung
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Lauren B. Alloy
- Department of PsychologyTemple UniversityPhiladelphiaPennsylvaniaUSA
| | - Sean Cain
- School of Psychological Sciences and Turner Institute for Brain and Mental HealthMonash UniversityMelbourneVictoriaAustralia
| | - Davide Dulcis
- UC San Diego Department of Psychiatry & Center for Circadian BiologyLa JollaCaliforniaUSA
| | - Bruno Etain
- Université de ParisINSERM UMR‐S 1144ParisFrance
| | - Benicio N. Frey
- Department Psychiatry and Behavioral NeuroscienceMcMaster UniversityHamiltonOntarioCanada
| | - Corrado Garbazza
- Centre for ChronobiologyPsychiatric Hospital of the University of Basel and Transfaculty Research Platform Molecular and Cognitive NeurosciencesUniversity of BaselBaselSwitzerland
| | - Kyle D. Ketchesin
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Dominic Landgraf
- Circadian Biology GroupDepartment of Molecular NeurobiologyClinic of Psychiatry and PsychotherapyUniversity HospitalLudwig Maximilian UniversityMunichGermany
| | - Heon‐Jeong Lee
- Department of Psychiatry and Chronobiology InstituteKorea UniversitySeoulSouth Korea
| | | | - Robin Nusslock
- Department of Psychology and Institute for Policy ResearchNorthwestern UniversityChicagoIllinoisUSA
| | - Alessandra Porcu
- UC San Diego Department of Psychiatry & Center for Circadian BiologyLa JollaCaliforniaUSA
| | | | - Philipp Ritter
- Clinic for Psychiatry and PsychotherapyCarl Gustav Carus University Hospital and Technical University of DresdenDresdenGermany
| | - Jan Scott
- Institute of NeuroscienceNewcastle UniversityNewcastleUK
| | - Daniel Smith
- Division of PsychiatryUniversity of EdinburghEdinburghUK
| | - Holly A. Swartz
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Greg Murray
- Centre for Mental HealthSwinburne University of TechnologyMelbourneVictoriaAustralia
| |
Collapse
|
15
|
Xue X, Zong W, Glausier JR, Kim SM, Shelton MA, Phan BN, Srinivasan C, Pfenning AR, Tseng GC, Lewis DA, Seney ML, Logan RW. Molecular rhythm alterations in prefrontal cortex and nucleus accumbens associated with opioid use disorder. Transl Psychiatry 2022; 12:123. [PMID: 35347109 PMCID: PMC8960783 DOI: 10.1038/s41398-022-01894-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 03/03/2022] [Accepted: 03/10/2022] [Indexed: 11/21/2022] Open
Abstract
Severe and persistent disruptions to sleep and circadian rhythms are common in people with opioid use disorder (OUD). Preclinical evidence suggests altered molecular rhythms in the brain modulate opioid reward and relapse. However, whether molecular rhythms are disrupted in the brains of people with OUD remained an open question, critical to understanding the role of circadian rhythms in opioid addiction. Using subjects' times of death as a marker of time of day, we investigated transcriptional rhythms in the brains of subjects with OUD compared to unaffected comparison subjects. We discovered rhythmic transcripts in both the dorsolateral prefrontal cortex (DLPFC) and nucleus accumbens (NAc), key brain areas involved in OUD, that were largely distinct between OUD and unaffected subjects. Fewer rhythmic transcripts were identified in DLPFC of subjects with OUD compared to unaffected subjects, whereas in the NAc, nearly double the number of rhythmic transcripts was identified in subjects with OUD. In NAc of subjects with OUD, rhythmic transcripts peaked either in the evening or near sunrise, and were associated with an opioid, dopamine, and GABAergic neurotransmission. Associations with altered neurotransmission in NAc were further supported by co-expression network analysis which identified OUD-specific modules enriched for transcripts involved in dopamine, GABA, and glutamatergic synaptic functions. Additionally, rhythmic transcripts in DLPFC and NAc of subjects with OUD were enriched for genomic loci associated with sleep-related GWAS traits, including sleep duration and insomnia. Collectively, our findings connect transcriptional rhythm changes in opioidergic, dopaminergic, GABAergic signaling in the human brain to sleep-related traits in opioid addiction.
Collapse
Affiliation(s)
- Xiangning Xue
- grid.21925.3d0000 0004 1936 9000Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15261 USA
| | - Wei Zong
- grid.21925.3d0000 0004 1936 9000Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15261 USA
| | - Jill R. Glausier
- grid.21925.3d0000 0004 1936 9000Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219 USA
| | - Sam-Moon Kim
- grid.21925.3d0000 0004 1936 9000Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219 USA ,grid.21925.3d0000 0004 1936 9000Center for Adolescent Reward, Rhythms, and Sleep, University of Pittsburgh, Pittsburgh, PA 15219 USA
| | - Micah A. Shelton
- grid.21925.3d0000 0004 1936 9000Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219 USA
| | - BaDoi N. Phan
- grid.147455.60000 0001 2097 0344Department of Computational Biology, Carnegie Mellon University, Pittsburgh, PA 15213 USA
| | - Chaitanya Srinivasan
- grid.147455.60000 0001 2097 0344Department of Computational Biology, Carnegie Mellon University, Pittsburgh, PA 15213 USA
| | - Andreas R. Pfenning
- grid.147455.60000 0001 2097 0344Department of Computational Biology, Carnegie Mellon University, Pittsburgh, PA 15213 USA ,grid.147455.60000 0001 2097 0344Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA 15213 USA
| | - George C. Tseng
- grid.21925.3d0000 0004 1936 9000Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15261 USA
| | - David A. Lewis
- grid.21925.3d0000 0004 1936 9000Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219 USA
| | - Marianne L. Seney
- grid.21925.3d0000 0004 1936 9000Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219 USA ,grid.21925.3d0000 0004 1936 9000Center for Adolescent Reward, Rhythms, and Sleep, University of Pittsburgh, Pittsburgh, PA 15219 USA
| | - Ryan W. Logan
- grid.189504.10000 0004 1936 7558Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118 USA ,grid.189504.10000 0004 1936 7558Center for Systems Neuroscience, Boston University, Boston, MA 02118 USA
| |
Collapse
|
16
|
Rapid-acting antidepressants and the circadian clock. Neuropsychopharmacology 2022; 47:805-816. [PMID: 34837078 PMCID: PMC8626287 DOI: 10.1038/s41386-021-01241-w] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 09/20/2021] [Accepted: 11/08/2021] [Indexed: 12/13/2022]
Abstract
A growing number of epidemiological and experimental studies has established that circadian disruption is strongly associated with psychiatric disorders, including major depressive disorder (MDD). This association is becoming increasingly relevant considering that modern lifestyles, social zeitgebers (time cues) and genetic variants contribute to disrupting circadian rhythms that may lead to psychiatric disorders. Circadian abnormalities associated with MDD include dysregulated rhythms of sleep, temperature, hormonal secretions, and mood which are modulated by the molecular clock. Rapid-acting antidepressants such as subanesthetic ketamine and sleep deprivation therapy can improve symptoms within 24 h in a subset of depressed patients, in striking contrast to conventional treatments, which generally require weeks for a full clinical response. Importantly, animal data show that sleep deprivation and ketamine have overlapping effects on clock gene expression. Furthermore, emerging data implicate the circadian system as a critical component involved in rapid antidepressant responses via several intracellular signaling pathways such as GSK3β, mTOR, MAPK, and NOTCH to initiate synaptic plasticity. Future research on the relationship between depression and the circadian clock may contribute to the development of novel therapeutic strategies for depression-like symptoms. In this review we summarize recent evidence describing: (1) how the circadian clock is implicated in depression, (2) how clock genes may contribute to fast-acting antidepressants, and (3) the mechanistic links between the clock genes driving circadian rhythms and neuroplasticity.
Collapse
|
17
|
Zhang Y, Jiang S, Liao F, Huang Z, Yang X, Zou Y, He X, Guo Q, Huang C. A transcriptomic analysis of neuropathic pain in the anterior cingulate cortex after nerve injury. Bioengineered 2022; 13:2058-2075. [PMID: 35030976 PMCID: PMC8973654 DOI: 10.1080/21655979.2021.2021710] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
The anterior cingulate cortex (ACC) is a core brain region processing pain emotion. In this study, we performed RNA sequencing analysis to reveal transcriptomic profiles of the ACC in a rat chronic constriction injury (CCI) model. A total of 1628 differentially expressed genes (DEGs) were identified by comparing sham-operated rats with rats of 12 hours, 1, 3, 7, and 14 days after surgery, respectively. Although these inflammatory-related DEGs were generally increased after CCI, different kinetics of time-series expression were observed with the development of neuropathic pain affection. Specifically, the expression of Ccl5, Cxcl9 and Cxcl13 continued to increase following CCI. The expression of Ccl2, Ccl3, Ccl4, Ccl6, and Ccl7 were initially upregulated after CCI and subsequently decreased after 12 hours. Similarly, the expression of Rac2, Cd68, Icam-1, Ptprc, Itgb2, and Fcgr2b increased after 12 hours but reduced after 1 day. However, the expression of the above genes increased again 7 days after CCI, when the neuropathic pain affection had developed. Furthermore, gene ontology analysis, Kyoto Encyclopedia of Genes and Genomes pathway enrichment and interaction network analyses further showed a high connectivity degree among these chemokine targeting genes. Similar expressional changes in these genes were found in the rat spinal dorsal horn responsible for nociception processing. Taken together, our results indicated chemokines and their targeting genes in the ACC may be differentially involved in the initiation and maintenance of neuropathic pain affection. These genes may be a target for not only the nociception but also the pain affection following nerve injury.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Shiwei Jiang
- Medical College of Xiangya, Central South University, Changsha, China
| | - Fei Liao
- Department of Anesthesiology, People's Hospital of Yuxi City, Yuxi, China
| | - Zhifeng Huang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Xin Yang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Yu Zou
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Xin He
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Qulian Guo
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Changsheng Huang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
18
|
Seney ML, Nestler EJ. Introduction to Special Issue: Insight Into Sex Differences in Neuropsychiatric Syndromes From Transcriptomic Analyses. Biol Psychiatry 2022; 91:3-5. [PMID: 34857105 PMCID: PMC8887677 DOI: 10.1016/j.biopsych.2021.09.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 09/08/2021] [Indexed: 11/28/2022]
Affiliation(s)
- Marianne L. Seney
- Department of Psychiatry and Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Eric J. Nestler
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|