1
|
Moradi S, Nouri M, Moradi MT, Khodarahmi R, Zarrabi M, Khazaie H. The mutual impacts of stem cells and sleep: opportunities for improved stem cell therapy. Stem Cell Res Ther 2025; 16:157. [PMID: 40158131 PMCID: PMC11954214 DOI: 10.1186/s13287-025-04235-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 02/17/2025] [Indexed: 04/01/2025] Open
Abstract
Sleep is an indispensable physiological function regulated by circadian rhythms, which influence the biological pathways and overall health of the body. Sleep is crucial for the maintenance and restoration of bodily systems, and disturbances can lead to various sleep disorders, which can impair both mental and physical health. Treatment options for these disorders encompass lifestyle modifications, psychotherapy, medications, and therapies such as light therapy and surgery. Not only sleep deprivation has a significant impact on essential organs, but it also influences various types of stem cells in the body. In this review, we explore the connection between sleep and various types of stem cells, highlighting how circadian rhythms regulate stem cell activities that are vital for tissue regeneration and homeostasis. Disruptions in sleep can hinder stem cell self-renewal, homing, proliferation, function, and differentiation, thereby affecting tissue regeneration and overall health. We also discuss how transplantation of stem cells and their products may help improve sleep disorders, how sleep quality affects stem cell behavior, and the implications for stem cell therapies. Notably, while certain stem cell transplantations can disrupt sleep, enhancing sleep quality may improve the efficacy of these therapies. Finally, stem cells can be utilized to model sleep disorders, offering valuable insights into their underlying mechanisms.
Collapse
Affiliation(s)
- Sharif Moradi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Masoumeh Nouri
- R&D Department, Royan Stem Cell Technology Co, Tehran, Iran
| | - Mohammad-Taher Moradi
- Sleep Disorders Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Reza Khodarahmi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Morteza Zarrabi
- R&D Department, Royan Stem Cell Technology Co, Tehran, Iran
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Habibolah Khazaie
- Sleep Disorders Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
2
|
Kohutek ZA, Caslin HL, Fehrenbach DJ, Heimlich JB, Brown JD, Madhur MS, Ferrell PB, Doran AC. Bone Marrow Niche in Cardiometabolic Disease: Mechanisms and Therapeutic Potential. Circ Res 2025; 136:325-353. [PMID: 39883790 PMCID: PMC11790260 DOI: 10.1161/circresaha.124.323778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2025]
Abstract
Cardiovascular and cardiometabolic diseases are leading causes of morbidity and mortality worldwide, driven in part by chronic inflammation. Emerging research suggests that the bone marrow microenvironment, or marrow niche, plays a critical role in both immune system regulation and disease progression. The bone marrow niche is essential for maintaining hematopoietic stem cells (HSCs) and orchestrating hematopoiesis. Under normal conditions, this niche ensures a return to immune homeostasis after acute stress. However, in the setting of inflammatory conditions such as those seen in cardiometabolic diseases, it becomes dysregulated, leading to enhanced myelopoiesis and immune activation. This review explores the reciprocal relationship between the bone marrow niche and cardiometabolic diseases, highlighting how alterations in the niche contribute to disease development and progression. The niche regulates HSCs through complex interactions with stromal cells, endothelial cells, and signaling molecules. However, in the setting of chronic diseases such as hypertension, atherosclerosis, and diabetes, inflammatory signals disrupt the balance between HSC self-renewal and differentiation, promoting the excessive production of proinflammatory myeloid cells that exacerbate the disease. Key mechanisms discussed include the effects of hyperlipidemia, hyperglycemia, and sympathetic nervous system activation on HSC proliferation and differentiation. Furthermore, the review emphasizes the role of epigenetic modifications and metabolic reprogramming in creating trained immunity, a phenomenon whereby HSCs acquire long-term proinflammatory characteristics that sustain disease states. Finally, we explore therapeutic strategies aimed at targeting the bone marrow niche to mitigate chronic inflammation and its sequelae. Novel interventions that modulate hematopoiesis and restore niche homeostasis hold promise for the treatment of cardiometabolic diseases. By interrupting the vicious cycle of inflammation and marrow dysregulation, such therapies may offer new avenues for reducing cardiovascular risk and improving patient outcomes.
Collapse
Affiliation(s)
- Zachary A. Kohutek
- Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Heather L. Caslin
- Department of Health and Human Performance, University of Houston, Houston, TX 77204, USA
| | - Daniel J. Fehrenbach
- Department of Medicine, Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - J. Brett Heimlich
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jonathan D. Brown
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Meena S. Madhur
- Department of Medicine, Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - P. Brent Ferrell
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University, Nashville, TN 37212, USA
| | - Amanda C. Doran
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University, Nashville, TN 37212, USA
| |
Collapse
|
3
|
Carpenter RS, Maryanovich M. Systemic and local regulation of hematopoietic homeostasis in health and disease. NATURE CARDIOVASCULAR RESEARCH 2024; 3:651-665. [PMID: 39196230 DOI: 10.1038/s44161-024-00482-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 04/24/2024] [Indexed: 08/29/2024]
Abstract
Hematopoietic stem cells (HSCs) generate all blood cell lineages responsible for tissue oxygenation, life-long hematopoietic homeostasis and immune protection. In adulthood, HSCs primarily reside in the bone marrow (BM) microenvironment, consisting of diverse cell types that constitute the stem cell 'niche'. The adaptability of the hematopoietic system is required to respond to the needs of the host, whether to maintain normal physiology or during periods of physical, psychosocial or environmental stress. Hematopoietic homeostasis is achieved by intricate coordination of systemic and local factors that orchestrate the function of HSCs throughout life. However, homeostasis is not a static process; it modulates HSC and progenitor activity in response to circadian rhythms coordinated by the central and peripheral nervous systems, inflammatory cues, metabolites and pathologic conditions. Here, we review local and systemic factors that impact hematopoiesis, focusing on the implications of aging, stress and cardiovascular disease.
Collapse
Affiliation(s)
- Randall S Carpenter
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Ruth L. and David S. Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Maria Maryanovich
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA.
- Ruth L. and David S. Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA.
- Cancer Dormancy and Tumor Microenvironment Institute, Albert Einstein College of Medicine, Bronx, NY, USA.
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
4
|
Martínez-Albert E, Lutz ND, Hübener R, Dimitrov S, Lange T, Born J, Besedovsky L. Sleep promotes T-cell migration towards CCL19 via growth hormone and prolactin signaling in humans. Brain Behav Immun 2024; 118:69-77. [PMID: 38369248 DOI: 10.1016/j.bbi.2024.02.021] [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: 08/23/2023] [Revised: 02/06/2024] [Accepted: 02/15/2024] [Indexed: 02/20/2024] Open
Abstract
Sleep strongly supports the formation of adaptive immunity, e.g., after vaccination. However, the underlying mechanisms remain largely obscure. Here we show in healthy humans that sleep compared to nocturnal wakefulness specifically promotes the migration of various T-cell subsets towards the chemokine CCL19, which is essential for lymph-node homing and, thus, for the initiation and maintenance of adaptive immune responses. Migration towards the inflammatory chemokine CCL5 remained unaffected. Incubating the cells with plasma from sleeping participants likewise increased CCL19-directed migration, an effect that was dependent on growth hormone and prolactin signaling. These findings show that sleep selectively promotes the lymph node homing potential of T cells by increasing hormonal release, and thus reveal a causal mechanism underlying the supporting effect of sleep on adaptive immunity in humans.
Collapse
Affiliation(s)
- Estefanía Martínez-Albert
- Institute of Medical Psychology and Behavioral Neurobiology, Eberhard Karls University of Tübingen, 72076 Tübingen, Germany; Institute of Medical Psychology, Ludwig-Maximilians-Universität München, 80336 Munich, Germany
| | - Nicolas D Lutz
- Institute of Medical Psychology and Behavioral Neurobiology, Eberhard Karls University of Tübingen, 72076 Tübingen, Germany; Institute of Medical Psychology, Ludwig-Maximilians-Universität München, 80336 Munich, Germany
| | - Robert Hübener
- Institute of Medical Psychology and Behavioral Neurobiology, Eberhard Karls University of Tübingen, 72076 Tübingen, Germany
| | - Stoyan Dimitrov
- Institute of Medical Psychology and Behavioral Neurobiology, Eberhard Karls University of Tübingen, 72076 Tübingen, Germany
| | - Tanja Lange
- Department of Rheumatology and Clinical Immunology, University of Lübeck, 23562 Lübeck, Germany; Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, 23562 Lübeck, Germany
| | - Jan Born
- Institute of Medical Psychology and Behavioral Neurobiology, Eberhard Karls University of Tübingen, 72076 Tübingen, Germany; Werner Reichardt Centre for Integrative Neuroscience, Eberhard Karls University of Tübingen, 72076 Tübingen, Germany
| | - Luciana Besedovsky
- Institute of Medical Psychology and Behavioral Neurobiology, Eberhard Karls University of Tübingen, 72076 Tübingen, Germany; Institute of Medical Psychology, Ludwig-Maximilians-Universität München, 80336 Munich, Germany.
| |
Collapse
|
5
|
Perioperative escape from dormancy of spontaneous micro-metastases: A role for malignant secretion of IL-6, IL-8, and VEGF, through adrenergic and prostaglandin signaling. Brain Behav Immun 2023; 109:175-187. [PMID: 36646396 DOI: 10.1016/j.bbi.2023.01.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 12/08/2022] [Accepted: 01/12/2023] [Indexed: 01/15/2023] Open
Abstract
We recently showed that a minimally-invasive removal of MDA-MB-231HM primary tumors (PTs) and elimination of their secreted factors (including IL-6, IL-8, VEGF, EGF, PDGF-aa, MIF, SerpinE1, and M-CSF), caused regression of spontaneous micro-metastases into a non-growing dormant state. To explore the underlying mechanisms and potential clinical ramifications of this phenomenon, we herein used the MDA-MB-231HM human breast cancer cell-line, in-vitro, and in vivo following orthotopic implantation in immune-deficient BALB/C nu/nu mice. Employing bioluminescence imaging, we found that adding laparotomy to minimally-invasive removal of the PT caused an outbreak of micro-metastases. However, perioperative β-adrenergic and COX-2 inhibition, using propranolol + etodolac, maintained metastatic dormancy following laparotomy. In-vitro, β-adrenergic agonists (epinephrine or metaproterenol) and prostaglandin-E2 markedly increased MDA-MB-231HM secretion of the pro-metastatic factors IL-6, IL-8, and VEGF, whereas cortisol reduced their secretion, effects that were maintained even 12 h after the washout of these agonists. In-vivo, laparotomy elevated IL-6 and IL-8 levels in both plasma and ex-vivo PT spontaneous secretion, whereas perioperative propranolol + etodolac administration blocked these effects. Similar trends were evident for EGF and MIF. Promoter-based bioinformatics analyses of excised PT transcriptomes implicated elevated NF-kB activity and reduced IRF1 activity in the gene regulatory effects of laparotomy, and these effects were inhibited by pre-surgical propranolol + etodolac. Taken together, our findings suggest a novel mechanism of post-operative metastatic outbreak, where surgery-induced adrenergic and prostanoid signaling increase the secretion of pro-metastatic factors, including IL-6, IL-8, and VEGF, from PT and possibly residual malignant tissue, and thereby prevent residual disease from entering dormancy.
Collapse
|
6
|
Li T, Ge H, Yang Q, Wang J, Yin Q, Wang H, Hou G. Oncogenic role of microRNA-19b-3p-mediated SOCS3 in glioma through activation of JAK-STAT pathway. Metab Brain Dis 2022; 38:945-960. [PMID: 36484970 DOI: 10.1007/s11011-022-01136-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 11/25/2022] [Indexed: 12/14/2022]
Abstract
The altered expression of microRNA (miRNA) has been implicated in glioma. Here, the current study aimed to clarify the oncogenic effects of miR-19b-3p on cellular processes of glioma and to elucidate the underlying mechanism associated with SOCS3 and the JAK-STAT signaling pathway. Differentially expressed genes related to glioma were initially identified via microarray analysis. Twenty-five glioma patients were selected for clinical data collection, while additional 12 patients with traumatic brain injuries were selected as controls. Cell senescence was assessed by β-galactosidase staining, proliferation by MTT assay and apoptosis by flow cytometry following gain- and/or loss-of-function of miR-19b-3p or SOCS3. Glioma xenograft mouse model was developed through subcutaneous injection to nude mice to provide evidence in vivo. The glioma patients exhibited overexpressed miR-19b-3p and poorly-expressed SOCS3. SOCS3 was identified as a target gene of miR-19b-3p through dual-luciferase reporter gene assay. miR-19b-3p repressed SOCS3 expression and activated the JAK-STAT signaling pathway. Furthermore, miR-19b-3p inhibition promoted apoptosis and senescence, and suppressed cell proliferation through inactivation of the JAK-STAT signaling pathway and up-regulation of SOCS3. The reported regulatory axis was validated in nude mice as evidenced by suppressed tumor growth. Taken together, this study demonstrates that miR-19b-3p facilitates glioma progression via activation of the JAK-STAT signaling pathway by targeting SOCS3, highlighting a novel therapeutic target for glioma treatment.
Collapse
Affiliation(s)
- Tao Li
- The Second Department of Neurosurgery, Affiliated Hospital of Hebei Engineering University, No. 81, Congtai Road, Congtai District, 056000, Handan, Hebei Province, P. R. China
| | - Hong Ge
- Personnel Department, Handan Psychiatric Hospital, 056000, Handan, P. R. China
| | - Qingyan Yang
- Department of Otolaryngology, Affiliated Hospital of Hebei Engineering University, 056000, Handan, P. R. China
| | - Junmei Wang
- The Fourth Department of Neurosurgery, Handan Central Hospital, 056000, Handan, P. R. China
| | - Qian Yin
- Department of Laboratory Medicine, Han Gang Hospital, 056000, Handan, P. R. China
| | - Hongbin Wang
- Department of Neurosurgery, Affiliated Hospital of Hebei Engineering University, 056000, Handan, P. R. China
| | - Gaolei Hou
- The Second Department of Neurosurgery, Affiliated Hospital of Hebei Engineering University, No. 81, Congtai Road, Congtai District, 056000, Handan, Hebei Province, P. R. China.
| |
Collapse
|
7
|
TNF signaling pathway-mediated microglial activation in the PFC underlies acute paradoxical sleep deprivation-induced anxiety-like behaviors in mice. Brain Behav Immun 2022; 100:254-266. [PMID: 34915154 DOI: 10.1016/j.bbi.2021.12.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/08/2021] [Accepted: 12/09/2021] [Indexed: 12/24/2022] Open
Abstract
Acute sleep deprivation is a common condition in modern life and increases anxiety symptoms in healthy individuals. The neuroinflammatory response induced by microglial activation could be an important contributing factor, but its underlying molecular mechanisms are still unclear. In the present study, we first found that acute paradoxical sleep deprivation (PSD) induced by the modified multiple platform method (MMPM) for 6 h led to anxiety-like behavior in mice, as verified by the open field test, elevated plus maze test, light-dark box test, and marble burying test. In addition, bioinformatic analysis suggested an important relationship between acute sleep deprivation and brain inflammatory signaling pathways. Key genes enriched in the TNF signaling pathway were confirmed to be altered during acute PSD by qPCR and Western blot analyses, including the upregulation of the prostaglandin-endoperoxide synthase 2 (Ptgs2) and suppressor of cytokine signaling 3 protein (Socs3) genes and the downregulation of the cysteine-aspartic acid protease 3 (Casp3) gene. Furthermore, we found that microglial cells in the prefrontal cortex (PFC) were activated with significant branch structure changes and that the cell body area was increased in the PSD model. Finally, we found that minocycline, a tetracycline with anti-inflammatory properties, may ameliorate the anxiogenic effect and microglial activation. Our study reveals significant correlations of anxiety-like behavior, microglial activation, and inflammation during acute PSD.
Collapse
|
8
|
Lange T, Luebber F, Grasshoff H, Besedovsky L. The contribution of sleep to the neuroendocrine regulation of rhythms in human leukocyte traffic. Semin Immunopathol 2022; 44:239-254. [PMID: 35041075 PMCID: PMC8901522 DOI: 10.1007/s00281-021-00904-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 11/03/2021] [Indexed: 12/12/2022]
Abstract
Twenty-four-hour rhythms in immune parameters and functions are robustly observed phenomena in biomedicine. Here, we summarize the important role of sleep and associated parameters on the neuroendocrine regulation of rhythmic immune cell traffic to different compartments, with a focus on human leukocyte subsets. Blood counts of "stress leukocytes" such as neutrophils, natural killer cells, and highly differentiated cytotoxic T cells present a rhythm with a daytime peak. It is mediated by morning increases in epinephrine, leading to a mobilization of these cells out of the marginal pool into the circulation following a fast, beta2-adrenoceptor-dependent inhibition of adhesive integrin signaling. In contrast, other subsets such as eosinophils and less differentiated T cells are redirected out of the circulation during daytime. This is mediated by stimulation of the glucocorticoid receptor following morning increases in cortisol, which promotes CXCR4-driven leukocyte traffic, presumably to the bone marrow. Hence, these cells show highest numbers in blood at night when cortisol levels are lowest. Sleep adds to these rhythms by actively suppressing epinephrine and cortisol levels. In addition, sleep increases levels of immunosupportive mediators, such as aldosterone and growth hormone, which are assumed to promote T-cell homing to lymph nodes, thus facilitating the initiation of adaptive immune responses during sleep. Taken together, sleep-wake behavior with its unique neuroendocrine changes regulates human leukocyte traffic with overall immunosupportive effects during nocturnal sleep. In contrast, integrin de-activation and redistribution of certain leukocytes to the bone marrow during daytime activity presumably serves immune regulation and homeostasis.
Collapse
Affiliation(s)
- Tanja Lange
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany. .,Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany.
| | - Finn Luebber
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany.,Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany.,Social Neuroscience Lab, University of Lübeck, Lübeck, Germany
| | - Hanna Grasshoff
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany.,Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
| | | |
Collapse
|
9
|
Deng Y, Julaiti A, Ran W, He Y. RETRACTED: Bone marrow mesenchymal stem cells-derived exosomal microRNA-19b-3p targets SOCS1 to facilitate progression of esophageal cancer. Life Sci 2021; 278:119491. [PMID: 33862112 DOI: 10.1016/j.lfs.2021.119491] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 04/06/2021] [Accepted: 04/09/2021] [Indexed: 12/21/2022]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the Editor-in-Chief. Concern was raised about the reliability of the Western blot results in Figs. 1D, 2K, 3C/F/G, 4B+H and 5B+H, which appear to have the same eyebrow shaped phenotype as many other publications tabulated here (https://docs.google.com/spreadsheets/d/149EjFXVxpwkBXYJOnOHb6RhAqT4a2llhj9LM60MBffM/edit#gid=0). The journal requested the corresponding author comment on these concerns and provide the raw data. However the authors were not able to satisfactorily fulfil this request and therefore the Editor-in-Chief decided to retract the article.
Collapse
Affiliation(s)
- Yanchao Deng
- Department of Thoracic Surgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang, China.
| | - Ainiwaer Julaiti
- Department of Thoracic Surgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang, China
| | - Wei Ran
- The First Clinical Medicine College of Xinjiang Medical University, Urumqi, 830054, Xinjiang, China
| | - Yao He
- The First Clinical Medicine College of Xinjiang Medical University, Urumqi, 830054, Xinjiang, China
| |
Collapse
|
10
|
Schiller M, Ben-Shaanan TL, Rolls A. Neuronal regulation of immunity: why, how and where? Nat Rev Immunol 2021; 21:20-36. [PMID: 32811994 DOI: 10.1038/s41577-020-0387-1] [Citation(s) in RCA: 133] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/18/2020] [Indexed: 02/07/2023]
Abstract
Neuroimmunology is one of the fastest-growing fields in the life sciences, and for good reason; it fills the gap between two principal systems of the organism, the nervous system and the immune system. Although both systems affect each other through bidirectional interactions, we focus here on one direction - the effects of the nervous system on immunity. First, we ask why is it beneficial to allow the nervous system any control over immunity? We evaluate the potential benefits to the immune system that arise by taking advantage of some of the brain's unique features, such as its capacity to integrate and synchronize physiological functions, its predictive capacity and its speed of response. Second, we explore how the brain communicates with the peripheral immune system, with a focus on the endocrine, sympathetic, parasympathetic, sensory and meningeal lymphatic systems. Finally, we examine where in the brain this immune information is processed and regulated. We chart a partial map of brain regions that may be relevant for brain-immune system communication, our goal being to introduce a conceptual framework for formulating new hypotheses to study these interactions.
Collapse
Affiliation(s)
- Maya Schiller
- Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Tamar L Ben-Shaanan
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
- Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Asya Rolls
- Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
11
|
Sharma A, Muresanu DF, Sahib S, Tian ZR, Castellani RJ, Nozari A, Lafuente JV, Buzoianu AD, Bryukhovetskiy I, Manzhulo I, Patnaik R, Wiklund L, Sharma HS. Concussive head injury exacerbates neuropathology of sleep deprivation: Superior neuroprotection by co-administration of TiO 2-nanowired cerebrolysin, alpha-melanocyte-stimulating hormone, and mesenchymal stem cells. PROGRESS IN BRAIN RESEARCH 2020; 258:1-77. [PMID: 33223033 DOI: 10.1016/bs.pbr.2020.09.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Sleep deprivation (SD) is common in military personnel engaged in combat operations leading to brain dysfunction. Military personnel during acute or chronic SD often prone to traumatic brain injury (TBI) indicating the possibility of further exacerbating brain pathology. Several lines of evidence suggest that in both TBI and SD alpha-melanocyte-stimulating hormone (α-MSH) and brain-derived neurotrophic factor (BDNF) levels decreases in plasma and brain. Thus, a possibility exists that exogenous supplement of α-MSH and/or BDNF induces neuroprotection in SD compounded with TBI. In addition, mesenchymal stem cells (MSCs) are very portent in inducing neuroprotection in TBI. We examined the effects of concussive head injury (CHI) in SD on brain pathology. Furthermore, possible neuroprotective effects of α-MSH, MSCs and neurotrophic factors treatment were explored in a rat model of SD and CHI. Rats subjected to 48h SD with CHI exhibited higher leakage of BBB to Evans blue and radioiodine compared to identical SD or CHI alone. Brain pathology was also exacerbated in SD with CHI group as compared to SD or CHI alone together with a significant reduction in α-MSH and BDNF levels in plasma and brain and enhanced level of tumor necrosis factor-alpha (TNF-α). Exogenous administration of α-MSH (250μg/kg) together with MSCs (1×106) and cerebrolysin (a balanced composition of several neurotrophic factors and active peptide fragments) (5mL/kg) significantly induced neuroprotection in SD with CHI. Interestingly, TiO2 nanowired delivery of α-MSH (100μg), MSCs, and cerebrolysin (2.5mL/kg) induced enhanced neuroprotection with higher levels of α-MSH and BDNF and decreased the TNF-α in SD with CHI. These observations are the first to show that TiO2 nanowired administration of α-MSH, MSCs and cerebrolysin induces superior neuroprotection following SD in CHI, not reported earlier. The clinical significance of our findings in light of the current literature is discussed.
Collapse
Affiliation(s)
- Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| | - Dafin F Muresanu
- Department of Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania; "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Seaab Sahib
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Z Ryan Tian
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Rudy J Castellani
- Department of Pathology, University of Maryland, Baltimore, MD, United States
| | - Ala Nozari
- Anesthesiology & Intensive Care, Massachusetts General Hospital, Boston, MA, United States
| | - José Vicente Lafuente
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Igor Bryukhovetskiy
- Department of Fundamental Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia; Laboratory of Pharmacology, National Scientific Center of Marine Biology, Far East Branch of the Russian Academy of Sciences, Vladivostok, Russia
| | - Igor Manzhulo
- Department of Fundamental Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia; Laboratory of Pharmacology, National Scientific Center of Marine Biology, Far East Branch of the Russian Academy of Sciences, Vladivostok, Russia
| | - Ranjana Patnaik
- Department of Biomaterials, School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
12
|
Ch R, Chevallier O, Elliott CT. Metabolomics reveal circadian control of cellular metabolism. Trends Analyt Chem 2020. [DOI: 10.1016/j.trac.2020.115986] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
13
|
Jiang Y, Rosborough BR, Chen J, Das S, Kitsios GD, McVerry BJ, Mallampalli RK, Lee JS, Ray A, Chen W, Ray P. Single cell RNA sequencing identifies an early monocyte gene signature in acute respiratory distress syndrome. JCI Insight 2020; 5:135678. [PMID: 32554932 PMCID: PMC7406263 DOI: 10.1172/jci.insight.135678] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 06/03/2020] [Indexed: 01/16/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) results from overwhelming pulmonary inflammation. Prior bulk RNA sequencing provided limited insights into ARDS pathogenesis. We used single cell RNA sequencing to probe ARDS at a higher resolution. PBMCs of patients with pneumonia and sepsis with early ARDS were compared with those of sepsis patients who did not develop ARDS. Monocyte clusters from ARDS patients revealed multiple distinguishing characteristics in comparison with monocytes from patients without ARDS, including downregulation of SOCS3 expression, accompanied by a proinflammatory signature with upregulation of multiple type I IFN-induced genes, especially in CD16+ cells. To generate an ARDS risk score, we identified upregulation of 29 genes in the monocytes of these patients, and 17 showed a similar profile in cells of patients in independent cohorts. Monocytes had increased expression of RAB11A, known to inhibit neutrophil efferocytosis; ATP2B1, a calcium pump that exports Ca2+ implicated in endothelial barrier disruption; and SPARC, associated with processing of procollagen to collagen. These data show that monocytes of ARDS patients upregulate expression of genes not just restricted to those associated with inflammation. Together, our findings identify molecules that are likely involved in ARDS pathogenesis that may inform biomarker and therapeutic development.
Collapse
Affiliation(s)
- Yale Jiang
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,School of Medicine, Tsinghua University, Beijing, China
| | - Brian R. Rosborough
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, and
| | - Jie Chen
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, and
| | - Sudipta Das
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, and
| | - Georgios D. Kitsios
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, and
| | - Bryan J. McVerry
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, and
| | - Rama K. Mallampalli
- Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University Wexner Medical Center, Davis Heart Lung Research Institute, Columbus, Ohio
| | - Janet S. Lee
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, and
| | - Anuradha Ray
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, and,Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Wei Chen
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Prabir Ray
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, and,Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
14
|
Hahn J, Günter M, Schuhmacher J, Bieber K, Pöschel S, Schütz M, Engelhardt B, Oster H, Sina C, Lange T, Autenrieth SE. Sleep enhances numbers and function of monocytes and improves bacterial infection outcome in mice. Brain Behav Immun 2020; 87:329-338. [PMID: 31904407 DOI: 10.1016/j.bbi.2020.01.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 12/15/2019] [Accepted: 01/01/2020] [Indexed: 01/21/2023] Open
Abstract
Sleep strongly impacts both humoral and cellular immunity; however, its acute effects on the innate immune defense against pathogens are unclear. Here, we elucidated in mice whether sleep affects the numbers and functions of innate immune cells and their defense against systemic bacterial infection. Sleep significantly increased numbers of classical monocytes in blood and spleen of mice that were allowed to sleep for six hours at the beginning of the normal resting phase compared to mice kept awake for the same time. The sleep-induced effect on classical monocytes was neither caused by alterations in corticosterone nor myelopoiesis, bone marrow egress or death of monocytes and did only partially involve Gαi-protein coupled receptors like chemokine receptor 2 (CCR2), but not the adhesion molecules intercellular adhesion molecule 1 (ICAM-1) or lymphocyte function-associated antigen 1 (LFA-1). Notably, sleep suppressed the expression of the clock gene Arntl in splenic monocytes and the sleep-induced increase in circulating classical monocytes was abrogated in Arntl-deficient animals, indicating that sleep is a prerequisite for clock-gene driven rhythmic trafficking of classical monocytes. Sleep also enhanced the production of reactive oxygen species by monocytes and neutrophils. Moreover, sleep profoundly reduced bacterial load in blood and spleen of mice that were allowed to sleep before systemic bacterial infection and consequently increased survival upon infection. These data provide the first evidence that sleep enhances numbers and function of innate immune cells and therewith strengthens early defense against bacterial pathogens.
Collapse
Affiliation(s)
- Julia Hahn
- Department of Internal Medicine II, University of Tübingen, 72076 Tübingen, Germany
| | - Manina Günter
- Department of Internal Medicine II, University of Tübingen, 72076 Tübingen, Germany
| | - Juliane Schuhmacher
- Department of Internal Medicine II, University of Tübingen, 72076 Tübingen, Germany
| | - Kristin Bieber
- Department of Internal Medicine II, University of Tübingen, 72076 Tübingen, Germany; Core Facility Flow Cytometry of the Medical Faculty Tübingen, University of Tübingen, 72076 Tübingen, Germany
| | - Simone Pöschel
- Department of Internal Medicine II, University of Tübingen, 72076 Tübingen, Germany; Core Facility Flow Cytometry of the Medical Faculty Tübingen, University of Tübingen, 72076 Tübingen, Germany
| | - Monika Schütz
- Institute for Medical Microbiology and Hygiene, University of Tübingen, 72076 Tübingen, Germany
| | - Britta Engelhardt
- Theodor Kocher Institute, University of Bern, 3012 Bern, Switzerland
| | - Henrik Oster
- Institute of Neurobiology, University of Lübeck, 23562 Lübeck, Germany
| | - Christian Sina
- Institute for Nutritional Medicine, University of Lübeck, 23562 Lübeck, Germany
| | - Tanja Lange
- Department of Rheumatology & Clinical Immunology, University of Lübeck, 23562 Lübeck, Germany
| | - Stella E Autenrieth
- Department of Internal Medicine II, University of Tübingen, 72076 Tübingen, Germany; Core Facility Flow Cytometry of the Medical Faculty Tübingen, University of Tübingen, 72076 Tübingen, Germany.
| |
Collapse
|
15
|
García-García A, Méndez-Ferrer S. The Autonomic Nervous System Pulls the Strings to Coordinate Circadian HSC Functions. Front Immunol 2020; 11:956. [PMID: 32508835 PMCID: PMC7251159 DOI: 10.3389/fimmu.2020.00956] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 04/23/2020] [Indexed: 11/16/2022] Open
Abstract
As for many other adult stem cells, the behavior of hematopoietic stem and progenitor cells (HSPCs) is subjected to circadian regulatory patterns. Multiple HSPC functions, such as proliferation, differentiation or trafficking exhibit time-dependent patterns that require a tight coordination to ensure daily blood cell production. The autonomic nervous system, together with circulating hormones, relay circadian signals from the central clock-the suprachiasmatic nucleus in the brain-to synchronize HSC niche physiology according to light/darkness cycles. Research over the last 20 years has revealed how specific neural signals modulate certain aspects of circadian HSC biology. However, only recently some studies have started to decipher the cellular and molecular mechanisms that orchestrate this complex regulation in a time-dependent fashion. Here we firstly review some of the recent key findings illustrating how different neural signals (catecholaminergic or cholinergic) regulate circadian HSC egress, homing, maintenance, proliferation, and differentiation. In particular, we highlight the critical role of different neurotransmitter receptors in the bone marrow microenvironment to channel these neural signals and regulate antagonistic processes according to circadian cues and organismal demands. Then, we discuss the potential biological meaning of HSC circadian regulation and its possible utility for clinical purposes. Finally, we offer our perspective on emerging concepts in HSC chronobiology.
Collapse
Affiliation(s)
- Andrés García-García
- Tissue Engineering, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Simón Méndez-Ferrer
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, Cambridge, United Kingdom
- National Health Service Blood and Transplant, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
16
|
Korin B, Avraham S, Azulay-Debby H, Farfara D, Hakim F, Rolls A. Short-term sleep deprivation in mice induces B cell migration to the brain compartment. Sleep 2020; 43:zsz222. [PMID: 31553459 PMCID: PMC7616588 DOI: 10.1093/sleep/zsz222] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 07/09/2019] [Indexed: 12/18/2022] Open
Abstract
Increasing evidence highlight the involvement of immune cells in brain activity and its dysfunction. The brain's immune compartment is a dynamic ensemble of cells that can fluctuate even in naive animals. However, the dynamics and factors that can affect the composition of immune cells in the naive brain are largely unknown. Here, we examined whether acute sleep deprivation can affect the brain's immune compartment (parenchyma, meninges, and choroid plexus). Using high-dimensional mass cytometry analysis, we broadly characterized the effects of short-term sleep deprivation on the immune composition in the mouse brain. We found that after 6 h of sleep deprivation, there was a significant increase in the abundance of B cells in the brain compartment. This effect can be accounted for, at least in part, by the elevated expression of the migration-related receptor, CXCR5, on B cells and its ligand, cxcl13, in the meninges following sleep deprivation. Thus, our study reveals that short-term sleep deprivation affects the brain's immune compartment, offering a new insight into how sleep disorders can affect brain function and potentially contribute to neurodegeneration and neuroinflammation.
Collapse
Affiliation(s)
- Ben Korin
- Department of Neuroscience, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
- Department of Immunology, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Shimrit Avraham
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Hilla Azulay-Debby
- Department of Neuroscience, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
- Department of Immunology, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Dorit Farfara
- Department of Neuroscience, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
- Department of Immunology, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Fahed Hakim
- Pediatric Pulmonary Unit, Rambam Health Care Campus, Haifa, Israel
- Cancer Research Center, EMMS Hospital, Nazareth, Israel
| | - Asya Rolls
- Department of Neuroscience, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
- Department of Immunology, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
- Technion Integrated Cancer Center (TICC), Technion - Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
17
|
Heller HC, Ruby NF. Functional Interactions Between Sleep and Circadian Rhythms in Learning and Learning Disabilities. Handb Exp Pharmacol 2019; 253:425-440. [PMID: 30443786 DOI: 10.1007/164_2018_176] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/13/2023]
Abstract
The propensity for sleep is timed by the circadian system. Many studies have shown that learning and memory performance is affected by circadian phase. And, of course it is well established that critical processes of memory consolidation occur during and depend on sleep. This chapter presents evidence that sleep and circadian rhythms do not just have separate influences on learning and memory that happen to coincide because of the circadian timing of sleep, but rather sleep and circadian systems have a critical functional interaction in the processes of memory consolidation. The evidence comes primarily from research on two models of learning disability: Down's syndrome model mice and Siberian hamsters. The Down's syndrome model mouse (Ts65Dn) has severe learning disability that has been shown to be due to GABAergic over-inhibition. Short-term, chronic therapies with GABAA antagonists restore learning ability in these mice long-term, but only if the antagonist treatments are given during the dark or sleep phase of the daily rhythm. The Siberian hamster is a model circadian animal except for the fact that a light treatment that gives the animal a phase advance on one day and a phase delay on the next day can result in total circadian arrhythmia for life. Once arrhythmic, the hamsters cannot learn. Learning, but not rhythmicity, is restored by short-term chronic treatment with GABA antagonists. Like many other species, if these hamsters are made arrhythmic by SCN lesion, their learning is unaffected. However, if made arrhythmic and learning disabled by the light treatment, subsequent lesions of their SCNs restore learning. SCN lesions also appear to restore learning in the Ts65Dn mice. The collective work on these two animal models of learning disability suggests that the circadian system modulates neuroplasticity. Our hypothesis is that a previously unrecognized function of the circadian system is to dampen neuroplasticity during the sleep phase to stabilize memory transcripts during their transfer to long-term memory. Thus, sleep and circadian systems have integrated roles to play in memory consolidation and do not just have separate but coincident influences on that process.
Collapse
Affiliation(s)
- H Craig Heller
- Biology Department, Stanford University, Stanford, CA, USA.
| | - Norman F Ruby
- Biology Department, Stanford University, Stanford, CA, USA
| |
Collapse
|
18
|
Golan K, Kollet O, Markus RP, Lapidot T. Daily light and darkness onset and circadian rhythms metabolically synchronize hematopoietic stem cell differentiation and maintenance: The role of bone marrow norepinephrine, tumor necrosis factor, and melatonin cycles. Exp Hematol 2019; 78:1-10. [PMID: 31494174 DOI: 10.1016/j.exphem.2019.08.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 08/29/2019] [Accepted: 08/31/2019] [Indexed: 12/16/2022]
Abstract
Hematopoietic stem and progenitor cells (HSPCs) are essential for daily mature blood cell production, host immunity, and osteoclast-mediated bone turnover. The timing at which stem cells give rise to mature blood and immune cells while maintaining the bone marrow (BM) reservoir of undifferentiated HSPCs and how these opposite tasks are synchronized are poorly understood. Previous studies revealed that daily light onset activates norepinephrine (NE)-induced BM CXCL12 downregulation, followed by CXCR4+ HSPC release to the circulation. Recently, we reported that daily light onset induces transient elevations of BM NE and tumor necrosis factor (TNF), which metabolically program BM HSPC differentiation and recruitment to replenish the blood. In contrast, darkness onset induces lower elevations of BM NE and TNF, activating melatonin production, which metabolically reprograms HSPCs, increasing their short- and long-term repopulation potential, and BM maintenance. How the functions of BM-retained HSPCs are influenced by daily light and darkness cycles and their clinical potential are further discussed.
Collapse
Affiliation(s)
- Karin Golan
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Orit Kollet
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Regina P Markus
- Laboratory of Chronopharmacology, Department of Physiology, Institute of Bioscience, University of São Paulo, São Paulo, Brazil
| | - Tsvee Lapidot
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
19
|
Rogers VE, Zhu S, Ancoli-Israel S, Liu L, Mandrell BN, Hinds PS. A pilot randomized controlled trial to improve sleep and fatigue in children with central nervous system tumors hospitalized for high-dose chemotherapy. Pediatr Blood Cancer 2019; 66:e27814. [PMID: 31081596 PMCID: PMC7416343 DOI: 10.1002/pbc.27814] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 04/29/2019] [Accepted: 04/30/2019] [Indexed: 12/22/2022]
Abstract
OBJECTIVES To determine whether a sleep intervention compared with standard of care (SOC) was successful in preserving nighttime sleep in children with central nervous system cancers hospitalized for high-dose chemotherapy (HDCT) and autologous stem cell rescue, and to explore associations between sleep and fatigue during treatment. METHODS An unblinded, randomized, controlled, multicomponent intervention (NCT00666614) including evidence-based cognitive and behavioral strategies to improve sleep was implemented in 33 children (age 4-12 years) and adolescents (age 13-19 years) during hospitalization. Children wore an actigraph to measure sleep and wake, and reported fatigue scores daily. Parents concurrently kept a sleep diary and reported fatigue scores for their children. RESULTS The mean age was 9.5 ± 3.9 years, 81.8% were white, and 60.6% were male. Sleep in all children was seriously disturbed throughout the study. Children in the intervention group maintained their longest nighttime sleep across the study, while it declined in children receiving SOC (P = 0.009 for interaction). There were few other differences in sleep between groups. Controlling for age and baseline fatigue, higher nighttime activity score, and lower percent sleep were significantly associated with higher next-day adolescent-reported fatigue (P < 0.05); longest sleep was significantly positively associated with next-day child-reported fatigue (P = 0.018). CONCLUSION In this sample of children undergoing HDCT, a multicomponent sleep intervention modestly preserved nighttime sleep duration, although overall sleep was poor in both groups. Sleep is an integral component of health, and may influence outcomes of children receiving HDCT. Further investigation into methods of preserving sleep in children undergoing intensive cancer therapy is warranted.
Collapse
Affiliation(s)
| | | | | | | | | | - Pamela S. Hinds
- Children’s National Health System, George Washington University, Washington, D.C
| |
Collapse
|
20
|
Chavez JS, Pietras EM. Hematopoietic Stem Cells Rock Around The Clock: Circadian Fate Control via TNF/ROS Signals. Cell Stem Cell 2019; 23:459-460. [PMID: 30290172 DOI: 10.1016/j.stem.2018.09.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Hematopoietic stem cell function is closely tied to circadian rhythms. In this issue of Cell Stem Cell, Golan et al. (2018) identify crosstalk between circadian hormone signals, the inflammatory cytokine TNF, and bone marrow macrophages as a key regulator of HSC proliferation, differentiation, and self-renewal in the bone marrow.
Collapse
Affiliation(s)
- James S Chavez
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Eric M Pietras
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Immunology & Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
21
|
Besedovsky L, Lange T, Haack M. The Sleep-Immune Crosstalk in Health and Disease. Physiol Rev 2019; 99:1325-1380. [PMID: 30920354 PMCID: PMC6689741 DOI: 10.1152/physrev.00010.2018] [Citation(s) in RCA: 787] [Impact Index Per Article: 131.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 10/29/2018] [Accepted: 10/29/2018] [Indexed: 02/08/2023] Open
Abstract
Sleep and immunity are bidirectionally linked. Immune system activation alters sleep, and sleep in turn affects the innate and adaptive arm of our body's defense system. Stimulation of the immune system by microbial challenges triggers an inflammatory response, which, depending on its magnitude and time course, can induce an increase in sleep duration and intensity, but also a disruption of sleep. Enhancement of sleep during an infection is assumed to feedback to the immune system to promote host defense. Indeed, sleep affects various immune parameters, is associated with a reduced infection risk, and can improve infection outcome and vaccination responses. The induction of a hormonal constellation that supports immune functions is one likely mechanism underlying the immune-supporting effects of sleep. In the absence of an infectious challenge, sleep appears to promote inflammatory homeostasis through effects on several inflammatory mediators, such as cytokines. This notion is supported by findings that prolonged sleep deficiency (e.g., short sleep duration, sleep disturbance) can lead to chronic, systemic low-grade inflammation and is associated with various diseases that have an inflammatory component, like diabetes, atherosclerosis, and neurodegeneration. Here, we review available data on this regulatory sleep-immune crosstalk, point out methodological challenges, and suggest questions open for future research.
Collapse
Affiliation(s)
- Luciana Besedovsky
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen , Tübingen , Germany ; Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School , Boston, Massachusetts ; and Department of Rheumatology and Clinical Immunology, University of Lübeck , Lübeck , Germany
| | - Tanja Lange
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen , Tübingen , Germany ; Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School , Boston, Massachusetts ; and Department of Rheumatology and Clinical Immunology, University of Lübeck , Lübeck , Germany
| | - Monika Haack
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen , Tübingen , Germany ; Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School , Boston, Massachusetts ; and Department of Rheumatology and Clinical Immunology, University of Lübeck , Lübeck , Germany
| |
Collapse
|
22
|
Sleep Matters: CD4 + T Cell Memory Formation and the Central Nervous System. Trends Immunol 2019; 40:674-686. [PMID: 31262652 DOI: 10.1016/j.it.2019.06.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 06/05/2019] [Accepted: 06/05/2019] [Indexed: 11/23/2022]
Abstract
The mechanisms of CD4+ T-cell memory formation in the immune system are debated. With the well-established concept of memory formation in the central nervous system (CNS), we propose that formation of CD4+ T-cell memory depends on the interaction of two different cell systems handling two types of stored information. First, information about antigen (event) and challenge (context) is taken up by antigen-presenting cells, as initial storage. Second, event and context information is transferred to CD4+ T cells. During activation, two categories of CD4+ T cell develop: effector CD4+ T cells, carrying event and context information, enabling them to efficiently focus their response to tissues under attack; and persisting CD4+ T cells, providing context-independent antigen-specific memories and long-term storage. This novel hypothesis is supported by the observation that mammalian sleep can improve both CNS and CD4+ T-cell memory.
Collapse
|
23
|
Timing's everything in light, location, and cells. Blood 2019; 133:186-187. [PMID: 30655302 DOI: 10.1182/blood-2018-11-883462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
24
|
Liu H, Chen A. Roles of sleep deprivation in cardiovascular dysfunctions. Life Sci 2019; 219:231-237. [PMID: 30630005 DOI: 10.1016/j.lfs.2019.01.006] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 12/14/2018] [Accepted: 01/06/2019] [Indexed: 01/09/2023]
Abstract
It is widely recognized that inadequate sleep is associated with multiple acute and chronic diseases and results in increased mortality and morbidity for cardiovascular diseases. In recent years, there has been increasing interest in sleep related investigations. Emerging evidence indicates that sleep deprivation changes the biological phenotypes of DNA, RNA and protein levels, but the underlying mechanisms are not clear. We summarized the current research on the detrimental roles of sleep deprivation on the heart and elucidated the underlying mechanisms of sleep deficiency to improve our understanding of sleep deprivation and the emerging strategies to target this process for therapeutic benefit.
Collapse
Affiliation(s)
- Haiqiong Liu
- Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, NO. 253, Gongye Avenue, 510282 Guangzhou, China; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, NO. 253, Gongye Avenue, 510282 Guangzhou, China; Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, China; Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, NO. 253, Gongye Avenue, 510282 Guangzhou, China
| | - Aihua Chen
- Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, NO. 253, Gongye Avenue, 510282 Guangzhou, China; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, NO. 253, Gongye Avenue, 510282 Guangzhou, China; Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, China; Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, NO. 253, Gongye Avenue, 510282 Guangzhou, China.
| |
Collapse
|
25
|
Clotaire DZJ, Du X, Wei Y, Yang D, Hua J. miR-19b-3p integrates Jak-Stat signaling pathway through Plzf to regulate self-renewal in dairy goat male germline stem cells. Int J Biochem Cell Biol 2018; 105:104-114. [PMID: 30393202 DOI: 10.1016/j.biocel.2018.10.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 10/19/2018] [Accepted: 10/25/2018] [Indexed: 12/27/2022]
Abstract
Jak-Stat pathway is the first pathway identified to stimulate spermatogonial stem cells (SSCs) self-renewal and maintenance activity. Recent studies have showed that stat3 a crucial gene implicated in this pathway can regulate self-renewal in male germline stem cell. In our previous study, we demonstrated that miR-19b-3p induces cell proliferation and reduces heterochromatin through Plzf which also regulates the balance between cell self-renewal and differentiation. Because miRNA can target several genes and to understand more about Plzf, a crucial transcription factor of SSCs, we performed microarray and found that miR-19b-3p integrate Jak-Stat through Plzf to regulate cell self-renewal. Our results demonstrated that miR-19b-3p induces Jak-Stat when Plzf is downregulated; overexpression of Plzf reversed the trend and shown an existence of feedback (-/+) between Plzf and GHR. The cell self-renewal markers CD49f, GFRα1, Oct4 and cKIT analyzed in the both groups miR-19b-3p and Plzf-overexpressing compared to their respective control confirm miR-19b-3p regulates cell pluripotency and self-renewal in goat male germline stem cells through Plzf. Together our finding revels that miR-19b-3p control Jak-Stat signaling through Plzf.
Collapse
Affiliation(s)
- Daguia Zambe John Clotaire
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi, 712100 China; Laboratoire des sciences Agronomiques et Biologiques pour le Développement (LASBAD), Faculty of Science, University of Bangui, P.O Box 908, Central Africa, Central African Republic
| | - Xiaomin Du
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi, 712100 China
| | - Yudong Wei
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi, 712100 China
| | - Donghui Yang
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi, 712100 China
| | - Jinlian Hua
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi, 712100 China.
| |
Collapse
|
26
|
Ben-Shaanan T, Schiller M, Rolls A. Studying brain-regulation of immunity with optogenetics and chemogenetics; A new experimental platform. Brain Behav Immun 2017; 65:1-8. [PMID: 27890661 DOI: 10.1016/j.bbi.2016.11.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 11/22/2016] [Accepted: 11/23/2016] [Indexed: 02/07/2023] Open
Abstract
The interactions between the brain and the immune system are bidirectional. Nevertheless, we have far greater understanding of how the immune system affects the brain than how the brain affects immunity. New technological developments such as optogenetics and chemogenetics (using DREADDs; Designer Receptors Exclusively Activated by Designer Drugs) can bridge this gap in our understanding, as they enable an unprecedented mechanistic and systemic analysis of the communication between the brain and the immune system. In this review, we discuss new experimental approaches for revealing neuronal circuits that can participate in regulation of immunity. In addition, we discuss methods, specifically optogenetics and chemogenetics, that enable targeted neuronal manipulation to reveal how different brain regions affect immunity. We describe how these techniques can be used as an experimental platform to address fundamental questions in psychoneuroimmunology and to understand how neuronal circuits associate with different psychological states can affect physiology.
Collapse
Affiliation(s)
- Tamar Ben-Shaanan
- Department of Immunology, Department of Neuroscience, Rappaport Medical School, Technion - Israel Institute of Technology, Haifa, Israel
| | - Maya Schiller
- Department of Immunology, Department of Neuroscience, Rappaport Medical School, Technion - Israel Institute of Technology, Haifa, Israel
| | - Asya Rolls
- Department of Immunology, Department of Neuroscience, Rappaport Medical School, Technion - Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
27
|
Ghosh S, Indracanti N, Joshi J, Indraganti PK. Rescuing Self: Transient Isolation and Autologous Transplantation of Bone Marrow Mitigates Radiation-Induced Hematopoietic Syndrome and Mortality in Mice. Front Immunol 2017; 8:1180. [PMID: 28993772 PMCID: PMC5622201 DOI: 10.3389/fimmu.2017.01180] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 09/06/2017] [Indexed: 01/19/2023] Open
Abstract
The inflamed bone marrow niche shortly after total body irradiation (TBI) is known to contribute to loss of hematopoietic stem cells in terms of their number and function. In this study, autologous bone marrow transfer (AL-BMT) was evaluated as a strategy for mitigating hematopoietic form of the acute radiation syndrome by timing the collection phase (2 h after irradiation) and reinfusion (24 h after irradiation) using mice as a model system. Collection of bone marrow (BM) cells (0.5 × 106 total marrow cells) 2 h after lethal TBI rescued different subclasses of hematopoietic stem and progenitor cells (HSPCs) from the detrimental inflammatory and damaging milieu in vivo. Cryopreservation of collected graft and its reinfusion 24 h after TBI significantly rescued mice from lethal effects of irradiation (65% survival against 0% in TBI group on day 30th) and hematopoietic depression. Transient hypometabolic state (HMS) induced 2 h after TBI effectively preserved the functional status of HSPCs and improved hematopoietic recovery even when BM was collected 8 h after TBI. Homing studies suggested that AL-BMT yielded similar percentages for different subsets of HSPCs when compared to syngeneic bone marrow transfer. The results suggest that the timing of collection, and reinfusion of graft is crucial for the success of AL-BMT.
Collapse
Affiliation(s)
- Subhajit Ghosh
- Division of Radiation Biosciences, Institute of Nuclear Medicine and Allied Sciences, Delhi, India.,S.N. Pradhan Centre for Neuroscience-University of Calcutta, Kolkata, India
| | - Namita Indracanti
- Division of Radiation Biosciences, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Jayadev Joshi
- Division of Radiation Biosciences, Institute of Nuclear Medicine and Allied Sciences, Delhi, India.,S.N. Pradhan Centre for Neuroscience-University of Calcutta, Kolkata, India
| | - Prem Kumar Indraganti
- Division of Radiation Biosciences, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| |
Collapse
|
28
|
Yaacoby-Bianu K, Hakim F. Sleep Disturbance and Cancer—Animal Models. CURRENT SLEEP MEDICINE REPORTS 2017. [DOI: 10.1007/s40675-017-0073-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
29
|
Chaix A, Zarrinpar A, Panda S. The circadian coordination of cell biology. J Cell Biol 2017; 215:15-25. [PMID: 27738003 PMCID: PMC5057284 DOI: 10.1083/jcb.201603076] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 09/21/2016] [Indexed: 02/07/2023] Open
Abstract
Chaix et al. review how cells generate circadian oscillations and how circadian clocks control cell biology. Circadian clocks are cell-autonomous timing mechanisms that organize cell functions in a 24-h periodicity. In mammals, the main circadian oscillator consists of transcription–translation feedback loops composed of transcriptional regulators, enzymes, and scaffolds that generate and sustain daily oscillations of their own transcript and protein levels. The clock components and their targets impart rhythmic functions to many gene products through transcriptional, posttranscriptional, translational, and posttranslational mechanisms. This, in turn, temporally coordinates many signaling pathways, metabolic activity, organelles’ structure and functions, as well as the cell cycle and the tissue-specific functions of differentiated cells. When the functions of these circadian oscillators are disrupted by age, environment, or genetic mutation, the temporal coordination of cellular functions is lost, reducing organismal health and fitness.
Collapse
Affiliation(s)
- Amandine Chaix
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Amir Zarrinpar
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037 Division of Gastroenterology, University of California, San Diego, La Jolla, CA 92093
| | - Satchidananda Panda
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037
| |
Collapse
|
30
|
McAlpine CS, Swirski FK. Circadian Influence on Metabolism and Inflammation in Atherosclerosis. Circ Res 2017; 119:131-41. [PMID: 27340272 DOI: 10.1161/circresaha.116.308034] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 03/11/2016] [Indexed: 11/16/2022]
Abstract
Many aspects of human health and disease display daily rhythmicity. The brain's suprachiasmic nucleus, which interprets recurring external stimuli, and autonomous molecular networks in peripheral cells together, set our biological circadian clock. Disrupted or misaligned circadian rhythms promote multiple pathologies including chronic inflammatory and metabolic diseases such as atherosclerosis. Here, we discuss studies suggesting that circadian fluctuations in the vessel wall and in the circulation contribute to atherogenesis. Data from humans and mice indicate that an impaired molecular clock, disturbed sleep, and shifting light-dark patterns influence leukocyte and lipid supply in the circulation and alter cellular behavior in atherosclerotic lesions. We propose that a better understanding of both local and systemic circadian rhythms in atherosclerosis will enhance clinical management, treatment, and public health policy.
Collapse
Affiliation(s)
- Cameron S McAlpine
- From the Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston.
| | - Filip K Swirski
- From the Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston
| |
Collapse
|
31
|
Boltze J, Nitzsche F, Jolkkonen J, Weise G, Pösel C, Nitzsche B, Wagner DC. Concise Review: Increasing the Validity of Cerebrovascular Disease Models and Experimental Methods for Translational Stem Cell Research. Stem Cells 2017; 35:1141-1153. [DOI: 10.1002/stem.2595] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 02/06/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Johannes Boltze
- Department of Translational Medicine and Cell Technology; Fraunhofer Research Institution for Marine Biotechnology and Cell Technology; Lübeck Germany
- Institute for Medical and Marine Biotechnology, University of Lübeck; Lübeck Germany
| | - Franziska Nitzsche
- Department of Cell Therapy; Fraunhofer Institute for Cell Therapy and Immunology; Leipzig Germany
- Department of Radiology; McGowan Institute for Regenerative Medicine, University of Pittsburgh; Pennsylvania USA
| | - Jukka Jolkkonen
- Department of Neurology; Institute of Clinical Medicine, University of Eastern Finland; Kuopio Finland
| | - Gesa Weise
- Department of Cell Therapy; Fraunhofer Institute for Cell Therapy and Immunology; Leipzig Germany
- Department of Neurology; University of Leipzig; Germany
| | - Claudia Pösel
- Department of Cell Therapy; Fraunhofer Institute for Cell Therapy and Immunology; Leipzig Germany
| | - Björn Nitzsche
- Department of Cell Therapy; Fraunhofer Institute for Cell Therapy and Immunology; Leipzig Germany
- Department of Nuclear Medicine; University Hospital Leipzig; Germany
| | - Daniel-Christoph Wagner
- Department of Cell Therapy; Fraunhofer Institute for Cell Therapy and Immunology; Leipzig Germany
- Institute of Pathology, University Medical Center Mainz; Germany
| |
Collapse
|
32
|
Lasisi DT, Shittu S, Meludu C, Salami A. Differential effects of total and partial sleep deprivation on salivary factors in Wistar rats. Arch Oral Biol 2017; 73:100-104. [PMID: 27744116 DOI: 10.1016/j.archoralbio.2016.09.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 08/25/2016] [Accepted: 09/13/2016] [Indexed: 12/22/2022]
|
33
|
Brown LA, Hasan S, Foster RG, Peirson SN. COMPASS: Continuous Open Mouse Phenotyping of Activity and Sleep Status. Wellcome Open Res 2016; 1:2. [PMID: 27976750 PMCID: PMC5140024 DOI: 10.12688/wellcomeopenres.9892.2] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background: Disruption of rhythms in activity and rest occur in many diseases, and provide an important indicator of healthy physiology and behaviour. However, outside the field of sleep and circadian rhythm research, these rhythmic processes are rarely measured due to the requirement for specialised resources and expertise. Until recently, the primary approach to measuring activity in laboratory rodents has been based on voluntary running wheel activity. By contrast, measuring sleep requires the use of electroencephalography (EEG), which involves invasive surgical procedures and time-consuming data analysis. Methods: Here we describe a simple, non-invasive system to measure home cage activity in mice based upon passive infrared (PIR) motion sensors. Careful calibration of this system will allow users to simultaneously assess sleep status in mice. The use of open-source tools and simple sensors keeps the cost and the size of data-files down, in order to increase ease of use and uptake. Results: In addition to providing accurate data on circadian activity parameters, here we show that extended immobility of >40 seconds provides a reliable indicator of sleep, correlating well with EEG-defined sleep (Pearson’s r >0.95, 4 mice). Conclusions: Whilst any detailed analysis of sleep patterns in mice will require EEG, behaviourally-defined sleep provides a valuable non-invasive means of simultaneously phenotyping both circadian rhythms and sleep. Whilst previous approaches have relied upon analysis of video data, here we show that simple motion sensors provide a cheap and effective alternative, enabling real-time analysis and longitudinal studies extending over weeks or even months. The data files produced are small, enabling easy deposition and sharing. We have named this system COMPASS - Continuous Open Mouse Phenotyping of Activity and Sleep Status. This simple approach is of particular value in phenotyping screens as well as providing an ideal tool to assess activity and rest cycles for non-specialists.
Collapse
Affiliation(s)
- Laurence A Brown
- Sleep and Circadian Neuroscience Institute (SCNi), Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Sibah Hasan
- Sleep and Circadian Neuroscience Institute (SCNi), Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Russell G Foster
- Sleep and Circadian Neuroscience Institute (SCNi), Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Stuart N Peirson
- Sleep and Circadian Neuroscience Institute (SCNi), Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
34
|
Brown LA, Hasan S, Foster RG, Peirson SN. COMPASS: Continuous Open Mouse Phenotyping of Activity and Sleep Status. Wellcome Open Res 2016. [PMID: 27976750 DOI: 10.12688/wellcomeopenres.9892.1] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Background Disruption of rhythms in activity and rest occur in many diseases, and provide an important indicator of healthy physiology and behaviour. However, outside the field of sleep and circadian rhythm research, these rhythmic processes are rarely measured due to the requirement for specialised resources and expertise. Until recently, the primary approach to measuring activity in laboratory rodents has been based on voluntary running wheel activity. By contrast, measuring sleep requires the use of electroencephalography (EEG), which involves invasive surgical procedures and time-consuming data analysis. Methods Here we describe a simple, non-invasive system to measure home cage activity in mice based upon passive infrared (PIR) motion sensors. Careful calibration of this system will allow users to simultaneously assess sleep status in mice. The use of open-source tools and simple sensors keeps the cost and the size of data-files down, in order to increase ease of use and uptake. Results In addition to providing accurate data on circadian activity parameters, here we show that extended immobility of >40 seconds provides a reliable indicator of sleep, correlating well with EEG-defined sleep (Pearson's r >0.95, 4 mice). Conclusions Whilst any detailed analysis of sleep patterns in mice will require EEG, behaviourally-defined sleep provides a valuable non-invasive means of simultaneously phenotyping both circadian rhythms and sleep. Whilst previous approaches have relied upon analysis of video data, here we show that simple motion sensors provide a cheap and effective alternative, enabling real-time analysis and longitudinal studies extending over weeks or even months. The data files produced are small, enabling easy deposition and sharing. We have named this system COMPASS - Continuous Open Mouse Phenotyping of Activity and Sleep Status. This simple approach is of particular value in phenotyping screens as well as providing an ideal tool to assess activity and rest cycles for non-specialists.
Collapse
Affiliation(s)
- Laurence A Brown
- Sleep and Circadian Neuroscience Institute (SCNi), Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Sibah Hasan
- Sleep and Circadian Neuroscience Institute (SCNi), Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Russell G Foster
- Sleep and Circadian Neuroscience Institute (SCNi), Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Stuart N Peirson
- Sleep and Circadian Neuroscience Institute (SCNi), Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
35
|
Puram RV, Kowalczyk MS, de Boer CG, Schneider RK, Miller PG, McConkey M, Tothova Z, Tejero H, Heckl D, Järås M, Chen MC, Li H, Tamayo A, Cowley GS, Rozenblatt-Rosen O, Al-Shahrour F, Regev A, Ebert BL. Core Circadian Clock Genes Regulate Leukemia Stem Cells in AML. Cell 2016; 165:303-16. [PMID: 27058663 PMCID: PMC4826477 DOI: 10.1016/j.cell.2016.03.015] [Citation(s) in RCA: 188] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Revised: 01/04/2016] [Accepted: 03/09/2016] [Indexed: 01/09/2023]
Abstract
Leukemia stem cells (LSCs) have the capacity to self-renew and propagate disease upon serial transplantation in animal models, and elimination of this cell population is required for curative therapies. Here, we describe a series of pooled, in vivo RNAi screens to identify essential transcription factors (TFs) in a murine model of acute myeloid leukemia (AML) with genetically and phenotypically defined LSCs. These screens reveal the heterodimeric, circadian rhythm TFs Clock and Bmal1 as genes required for the growth of AML cells in vitro and in vivo. Disruption of canonical circadian pathway components produces anti-leukemic effects, including impaired proliferation, enhanced myeloid differentiation, and depletion of LSCs. We find that both normal and malignant hematopoietic cells harbor an intact clock with robust circadian oscillations, and genetic knockout models reveal a leukemia-specific dependence on the pathway. Our findings establish a role for the core circadian clock genes in AML.
Collapse
Affiliation(s)
- Rishi V Puram
- Department of Medicine, Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of Harvard University and MIT, Cambridge, MA 02142, USA
| | | | - Carl G de Boer
- Broad Institute of Harvard University and MIT, Cambridge, MA 02142, USA
| | - Rebekka K Schneider
- Department of Medicine, Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Peter G Miller
- Department of Medicine, Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Marie McConkey
- Department of Medicine, Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Zuzana Tothova
- Department of Medicine, Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of Harvard University and MIT, Cambridge, MA 02142, USA; Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Héctor Tejero
- Translational Bioinformatics Unit, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain
| | - Dirk Heckl
- Department of Medicine, Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Pediatric Hematology and Oncology, Hanover Medical School, Hanover 30625, Germany
| | - Marcus Järås
- Department of Medicine, Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Clinical Genetics, Lund University, Lund 22184, Sweden
| | - Michelle C Chen
- Department of Medicine, Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Hubo Li
- Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA 02115, USA; Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Alfred Tamayo
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Glenn S Cowley
- Broad Institute of Harvard University and MIT, Cambridge, MA 02142, USA
| | | | - Fatima Al-Shahrour
- Department of Medicine, Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Translational Bioinformatics Unit, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain
| | - Aviv Regev
- Broad Institute of Harvard University and MIT, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Benjamin L Ebert
- Department of Medicine, Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of Harvard University and MIT, Cambridge, MA 02142, USA; Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|