1
|
Xu S, Jia M, Guo J, He J, Chen X, Xu Y, Hu W, Wu D, Wu C, Ji X. Ticking Brain: Circadian Rhythm as a New Target for Cerebroprotection. Stroke 2024; 55:2385-2396. [PMID: 39011642 DOI: 10.1161/strokeaha.124.046684] [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] [Indexed: 07/17/2024]
Abstract
Circadian rhythm is a master process observed in nearly every type of cell throughout the body, and it macroscopically regulates daily physiology. Recent clinical trials have revealed the effects of circadian variation on the incidence, pathophysiological processes, and prognosis of acute ischemic stroke. Furthermore, core clock genes, the cell-autonomous pacemakers of the circadian rhythm, affect the neurovascular unit-composing cells in a nonparallel manner after the same pathophysiological processes of ischemia/reperfusion. In this review, we discuss the influence of circadian rhythms and clock genes on each type of neurovascular unit cell in the pathophysiological processes of acute ischemic stroke.
Collapse
Affiliation(s)
- Shuaili Xu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders (S.X., X.J.), Capital Medical University, Beijing, China
- China-America Institute of Neuroscience, Xuanwu Hospital (S.X., J.G., J.H., X.C., Y.X., W.H., D.W., X.J.), Capital Medical University, Beijing, China
| | - Milan Jia
- Department of Neurology, Xuanwu Hospital (M.J., X.C., Y.X., W.H., C.W., X.J.), Capital Medical University, Beijing, China
| | - Jiaqi Guo
- China-America Institute of Neuroscience, Xuanwu Hospital (S.X., J.G., J.H., X.C., Y.X., W.H., D.W., X.J.), Capital Medical University, Beijing, China
| | - Jiachen He
- China-America Institute of Neuroscience, Xuanwu Hospital (S.X., J.G., J.H., X.C., Y.X., W.H., D.W., X.J.), Capital Medical University, Beijing, China
| | - Xi Chen
- Department of Neurology, Xuanwu Hospital (M.J., X.C., Y.X., W.H., C.W., X.J.), Capital Medical University, Beijing, China
- China-America Institute of Neuroscience, Xuanwu Hospital (S.X., J.G., J.H., X.C., Y.X., W.H., D.W., X.J.), Capital Medical University, Beijing, China
| | - Yi Xu
- Department of Neurology, Xuanwu Hospital (M.J., X.C., Y.X., W.H., C.W., X.J.), Capital Medical University, Beijing, China
- China-America Institute of Neuroscience, Xuanwu Hospital (S.X., J.G., J.H., X.C., Y.X., W.H., D.W., X.J.), Capital Medical University, Beijing, China
| | - Wenbo Hu
- Department of Neurology, Xuanwu Hospital (M.J., X.C., Y.X., W.H., C.W., X.J.), Capital Medical University, Beijing, China
- China-America Institute of Neuroscience, Xuanwu Hospital (S.X., J.G., J.H., X.C., Y.X., W.H., D.W., X.J.), Capital Medical University, Beijing, China
| | - Di Wu
- China-America Institute of Neuroscience, Xuanwu Hospital (S.X., J.G., J.H., X.C., Y.X., W.H., D.W., X.J.), Capital Medical University, Beijing, China
| | - Chuanjie Wu
- Department of Neurology, Xuanwu Hospital (M.J., X.C., Y.X., W.H., C.W., X.J.), Capital Medical University, Beijing, China
| | - Xunming Ji
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders (S.X., X.J.), Capital Medical University, Beijing, China
- Department of Neurology, Xuanwu Hospital (M.J., X.C., Y.X., W.H., C.W., X.J.), Capital Medical University, Beijing, China
- China-America Institute of Neuroscience, Xuanwu Hospital (S.X., J.G., J.H., X.C., Y.X., W.H., D.W., X.J.), Capital Medical University, Beijing, China
- Department of Neurosurgery, Xuanwu Hospital (X.J.), Capital Medical University, Beijing, China
| |
Collapse
|
2
|
Lawrence JH, Patel A, King MW, Nadarajah CJ, Daneman R, Musiek ES. Microglia drive diurnal variation in susceptibility to inflammatory blood-brain barrier breakdown. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.10.588924. [PMID: 38645230 PMCID: PMC11030435 DOI: 10.1101/2024.04.10.588924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
The blood-brain barrier (BBB) is critical for maintaining brain homeostasis but is susceptible to inflammatory dysfunction. Permeability of the BBB to lipophilic molecules shows circadian variation due to rhythmic transporter expression, while basal permeability to polar molecules is non-rhythmic. Whether daily timing influences BBB permeability in response to inflammation is unknown. Here, we induced systemic inflammation through repeated lipopolysaccharide (LPS) injections either in the morning (ZT1) or evening (ZT13) under standard lighting conditions, then examined BBB permeability to a polar molecule, sodium fluorescein. We observed clear diurnal variation in inflammatory BBB permeability, with a striking increase in paracellular leak across the BBB specifically following evening LPS injection. Evening LPS led to persisting glia activation and inflammation in the brain that was not observed in the periphery. The exaggerated evening neuroinflammation and BBB disruption were suppressed by microglial depletion or through keeping mice in constant darkness. Our data show that diurnal rhythms in microglial inflammatory responses to LPS drive daily variability in BBB breakdown and reveals time-of-day as a key regulator of inflammatory BBB disruption.
Collapse
|
3
|
Abstract
The blood-brain barrier (BBB) is a critical interface separating the central nervous system from the peripheral circulation, ensuring brain homeostasis and function. Recent research has unveiled a profound connection between the BBB and circadian rhythms, the endogenous oscillations synchronizing biological processes with the 24-hour light-dark cycle. This review explores the significance of circadian rhythms in the context of BBB functions, with an emphasis on substrate passage through the BBB. Our discussion includes efflux transporters and the molecular timing mechanisms that regulate their activities. A significant focus of this review is the potential implications of chronotherapy, leveraging our knowledge of circadian rhythms for improving drug delivery to the brain. Understanding the temporal changes in BBB can lead to optimized timing of drug administration, to enhance therapeutic efficacy for neurological disorders while reducing side effects. By elucidating the interplay between circadian rhythms and drug transport across the BBB, this review offers insights into innovative therapeutic interventions.
Collapse
Affiliation(s)
- Mari Kim
- Cell Biology Department, Emory University, Atlanta, GA, USA (M.K., S.L.Z.)
| | - Richard F Keep
- Neurosurgery, University of Michigan, Ann Arbor, MI, USA (R.F.K.)
| | - Shirley L Zhang
- Cell Biology Department, Emory University, Atlanta, GA, USA (M.K., S.L.Z.)
| |
Collapse
|
4
|
Abstract
The brain is a complex organ, fundamentally changing across the day to perform basic functions like sleep, thought, and regulating whole-body physiology. This requires a complex symphony of nutrients, hormones, ions, neurotransmitters and more to be properly distributed across the brain to maintain homeostasis throughout 24 hours. These solutes are distributed both by the blood and by cerebrospinal fluid. Cerebrospinal fluid contents are distinct from the general circulation because of regulation at brain barriers including the choroid plexus, glymphatic system, and blood-brain barrier. In this review, we discuss the overlapping circadian (≈24-hour) rhythms in brain fluid biology and at the brain barriers. Our goal is for the reader to gain both a fundamental understanding of brain barriers alongside an understanding of the interactions between these fluids and the circadian timing system. Ultimately, this review will provide new insight into how alterations in these finely tuned clocks may lead to pathology.
Collapse
Affiliation(s)
- Velia S Vizcarra
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Ryann M Fame
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Lauren M Hablitz
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| |
Collapse
|
5
|
Skapetze L, Owino S, Lo EH, Arai K, Merrow M, Harrington M. Rhythms in barriers and fluids: Circadian clock regulation in the aging neurovascular unit. Neurobiol Dis 2023; 181:106120. [PMID: 37044366 DOI: 10.1016/j.nbd.2023.106120] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/27/2023] [Accepted: 04/07/2023] [Indexed: 04/14/2023] Open
Abstract
The neurovascular unit is where two very distinct physiological systems meet: The central nervous system (CNS) and the blood. The permeability of the barriers separating these systems is regulated by time, including both the 24 h circadian clock and the longer processes of aging. An endogenous circadian rhythm regulates the transport of molecules across the blood-brain barrier and the circulation of the cerebrospinal fluid and the glymphatic system. These fluid dynamics change with time of day, and with age, and especially in the context of neurodegeneration. Factors may differ depending on brain region, as can be highlighted by consideration of circadian regulation of the neurovascular niche in white matter. As an example of a potential target for clinical applications, we highlight chaperone-mediated autophagy as one mechanism at the intersection of circadian dysregulation, aging and neurodegenerative disease. In this review we emphasize key areas for future research.
Collapse
Affiliation(s)
- Lea Skapetze
- Institute of Medical Psychology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Sharon Owino
- Neuroscience Program, Smith College, Northampton, MA 01060, United States of America
| | - Eng H Lo
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ken Arai
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Martha Merrow
- Institute of Medical Psychology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Mary Harrington
- Neuroscience Program, Smith College, Northampton, MA 01060, United States of America.
| |
Collapse
|
6
|
Contreras EG, Klämbt C. The Drosophila blood-brain barrier emerges as a model for understanding human brain diseases. Neurobiol Dis 2023; 180:106071. [PMID: 36898613 DOI: 10.1016/j.nbd.2023.106071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/24/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
The accurate regulation of the microenvironment within the nervous system is one of the key features characterizing complex organisms. To this end, neural tissue has to be physically separated from circulation, but at the same time, mechanisms must be in place to allow controlled transport of nutrients and macromolecules into and out of the brain. These roles are executed by cells of the blood-brain barrier (BBB) found at the interface of circulation and neural tissue. BBB dysfunction is observed in several neurological diseases in human. Although this can be considered as a consequence of diseases, strong evidence supports the notion that BBB dysfunction can promote the progression of brain disorders. In this review, we compile the recent evidence describing the contribution of the Drosophila BBB to the further understanding of brain disease features in human patients. We discuss the function of the Drosophila BBB during infection and inflammation, drug clearance and addictions, sleep, chronic neurodegenerative disorders and epilepsy. In summary, this evidence suggests that the fruit fly, Drosophila melanogaster, can be successfully employed as a model to disentangle mechanisms underlying human diseases.
Collapse
Affiliation(s)
- Esteban G Contreras
- University of Münster, Institute of Neuro- and Behavioral Biology, Badestr. 9, Münster, Germany.
| | - Christian Klämbt
- University of Münster, Institute of Neuro- and Behavioral Biology, Badestr. 9, Münster, Germany.
| |
Collapse
|
7
|
Schurhoff N, Toborek M. Circadian rhythms in the blood-brain barrier: impact on neurological disorders and stress responses. Mol Brain 2023; 16:5. [PMID: 36635730 PMCID: PMC9835375 DOI: 10.1186/s13041-023-00997-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 01/03/2023] [Indexed: 01/14/2023] Open
Abstract
Circadian disruption has become more prevalent in society due to the increase in shift work, sleep disruption, blue light exposure, and travel via different time zones. The circadian rhythm is a timed transcription-translation feedback loop with positive regulators, BMAL1 and CLOCK, that interact with negative regulators, CRY and PER, to regulate both the central and peripheral clocks. This review highlights the functions of the circadian rhythm, specifically in the blood-brain barrier (BBB), during both healthy and pathological states. The BBB is a highly selective dynamic interface composed of CNS endothelial cells, astrocytes, pericytes, neurons, and microglia that form the neurovascular unit (NVU). Circadian rhythms modulate BBB integrity through regulating oscillations of tight junction proteins, assisting in functions of the NVU, and modulating transporter functions. Circadian disruptions within the BBB have been observed in stress responses and several neurological disorders, including brain metastasis, epilepsy, Alzheimer's disease, and Parkinson's disease. Further understanding of these interactions may facilitate the development of improved treatment options and preventative measures.
Collapse
Affiliation(s)
- Nicolette Schurhoff
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Suite 528, 1011 NW 15th Street, Miami, FL, 33155, USA
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Suite 528, 1011 NW 15th Street, Miami, FL, 33155, USA.
- Institute of Physiotherapy and Health Sciences, The Jerzy Kukuczka Academy of Physical Education, 40-065, Katowice, Poland.
| |
Collapse
|
8
|
Abstract
Leptin for over 25 years has been a central theme in the study of appetite, obesity, and starvation. As the major site of leptin production is peripheral, and the site of action of greatest interest is the hypothalamus, how leptin accesses the central nervous system (CNS) and crosses the blood-brain barrier (BBB) has been of great interest. We review here the ongoing research that addresses fundamental questions such as the sites of leptin resistances in obesity and other conditions, the causes of resistances and their relations to one another, the three barrier sites of entry into the CNS, why recent studies using suprapharmacological doses cannot address these questions but give insight into nonsaturable entry of leptin into the CNS, and how that might be useful in using leptin therapeutically. The current status of the controversy of whether the short form of the leptin receptor acts as the BBB leptin transporter and how obesity may transform leptin transport is reviewed. Review of these and other topics summarizes in a new appreciation of what leptin may have actually evolved to do and what physiological role leptin resistance may play. © 2021 American Physiological Society. Compr Physiol 11:1-19, 2021.
Collapse
Affiliation(s)
- William A Banks
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington, USA.,Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
9
|
Zhang SL, Lahens NF, Yue Z, Arnold DM, Pakstis PP, Schwarz JE, Sehgal A. A circadian clock regulates efflux by the blood-brain barrier in mice and human cells. Nat Commun 2021; 12:617. [PMID: 33504784 PMCID: PMC7841146 DOI: 10.1038/s41467-020-20795-9] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 12/18/2020] [Indexed: 01/30/2023] Open
Abstract
The blood-brain barrier (BBB) is critical for neural function. We report here circadian regulation of the BBB in mammals. Efflux of xenobiotics by the BBB oscillates in mice, with highest levels during the active phase and lowest during the resting phase. This oscillation is abrogated in circadian clock mutants. To elucidate mechanisms of circadian regulation, we profiled the transcriptome of brain endothelial cells; interestingly, we detected limited circadian regulation of transcription, with no evident oscillations in efflux transporters. We recapitulated the cycling of xenobiotic efflux using a human microvascular endothelial cell line to find that the molecular clock drives cycling of intracellular magnesium through transcriptional regulation of TRPM7, which appears to contribute to the rhythm in efflux. Our findings suggest that considering circadian regulation may be important when therapeutically targeting efflux transporter substrates to the CNS.
Collapse
Affiliation(s)
- Shirley L Zhang
- Chronobiology and Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
- Howard Hughes Medical Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| | - Nicholas F Lahens
- Institute for Translational Medicine and Therapeutics (ITMAT), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhifeng Yue
- Chronobiology and Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Howard Hughes Medical Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Denice M Arnold
- Chronobiology and Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Peter P Pakstis
- Chronobiology and Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Jessica E Schwarz
- Chronobiology and Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Amita Sehgal
- Chronobiology and Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
- Howard Hughes Medical Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
10
|
MCH Neurons Regulate Permeability of the Median Eminence Barrier. Neuron 2020; 107:306-319.e9. [PMID: 32407670 PMCID: PMC7383232 DOI: 10.1016/j.neuron.2020.04.020] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 03/06/2020] [Accepted: 04/20/2020] [Indexed: 01/17/2023]
Abstract
Melanin-concentrating hormone (MCH)-expressing neurons are key regulators of energy and glucose homeostasis. Here, we demonstrate that they provide dense projections to the median eminence (ME) in close proximity to tanycytes and fenestrated vessels. Chemogenetic activation of MCH neurons as well as optogenetic stimulation of their projections in the ME enhance permeability of the ME by increasing fenestrated vascular loops and enhance leptin action in the arcuate nucleus of the hypothalamus (ARC). Unbiased phosphoRiboTrap-based assessment of cell activation upon chemogenetic MCH neuron activation reveals MCH-neuron-dependent regulation of endothelial cells. MCH neurons express the vascular endothelial growth factor A (VEGFA), and blocking VEGF-R signaling attenuates the leptin-sensitizing effect of MCH neuron activation. Our experiments reveal that MCH neurons directly regulate permeability of the ME barrier, linking the activity of energy state and sleep regulatory neurons to the regulation of hormone accessibility to the ARC. MCH neurons provide dense projections to the median eminence MCH neuron activation promotes permeability of the median eminence barrier MCH neuron activation enhances microvessel fenestration in the ME MCH neuron activation enhances leptin action in the arcuate nucleus
Collapse
|
11
|
Abstract
Studies have linked obesity, metabolic syndrome, type 2 diabetes, cardiovascular disease (CVD), nonalcoholic fatty liver disease (NAFLD) and dementia. Their relationship to the incidence and progression of these disease states suggests an interconnected pathogenesis involving chronic low-grade inflammation and oxidative stress. Metabolic syndrome represents comorbidities of central obesity, insulin resistance, dyslipidemia, hypertension and hyperglycemia associated with increased risk of type 2 diabetes, NAFLD, atherosclerotic CVD and neurodegenerative disease. As the socioeconomic burden for these diseases has grown signficantly with an increasing elderly population, new and alternative pharmacologic solutions for these cardiometabolic diseases are required. Adipose tissue, skeletal muscle and liver are central endocrine organs that regulate inflammation, energy and metabolic homeostasis, and the neuroendocrine axis through synthesis and secretion of adipokines, myokines, and hepatokines, respectively. These organokines affect each other and communicate through various endocrine, paracrine and autocrine pathways. The ultimate goal of this review is to provide a comprehensive understanding of organ crosstalk. This will include the roles of novel organokines in normal physiologic regulation and their pathophysiological effect in obesity, metabolic syndrome, type 2 diabetes, CVD, NAFLD and neurodegenerative disorders.
Collapse
Affiliation(s)
- Hye Soo Chung
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Hallym University, Seoul, South Korea
| | - Kyung Mook Choi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Korea University, Seoul, South Korea.
| |
Collapse
|
12
|
Cuddapah VA, Zhang SL, Sehgal A. Regulation of the Blood-Brain Barrier by Circadian Rhythms and Sleep. Trends Neurosci 2019; 42:500-510. [PMID: 31253251 PMCID: PMC6602072 DOI: 10.1016/j.tins.2019.05.001] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 04/27/2019] [Accepted: 05/01/2019] [Indexed: 01/09/2023]
Abstract
The blood-brain barrier (BBB) is an evolutionarily conserved, structural, and functional separation between circulating blood and the central nervous system (CNS). By controlling permeability into and out of the nervous system, the BBB has a critical role in the precise regulation of neural processes. Here, we review recent studies demonstrating that permeability at the BBB is dynamically controlled by circadian rhythms and sleep. An endogenous circadian rhythm in the BBB controls transporter function, regulating permeability across the BBB. In addition, sleep promotes the clearance of metabolites along the BBB, as well as endocytosis across the BBB. Finally, we highlight the implications of this regulation for diseases, including epilepsy.
Collapse
Affiliation(s)
- Vishnu Anand Cuddapah
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Center for Sleep and Circadian Neurobiology, Chronobiology Program, and Howard Hughes Medical Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shirley L Zhang
- Center for Sleep and Circadian Neurobiology, Chronobiology Program, and Howard Hughes Medical Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Amita Sehgal
- Center for Sleep and Circadian Neurobiology, Chronobiology Program, and Howard Hughes Medical Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
13
|
Circadian Clocks and Sleep: Impact of Rhythmic Metabolism and Waste Clearance on the Brain. Trends Neurosci 2018; 41:677-688. [DOI: 10.1016/j.tins.2018.07.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 06/20/2018] [Accepted: 07/10/2018] [Indexed: 12/13/2022]
|
14
|
Zhang SL, Yue Z, Arnold DM, Artiushin G, Sehgal A. A Circadian Clock in the Blood-Brain Barrier Regulates Xenobiotic Efflux. Cell 2018; 173:130-139.e10. [PMID: 29526461 PMCID: PMC5866247 DOI: 10.1016/j.cell.2018.02.017] [Citation(s) in RCA: 146] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Revised: 12/22/2017] [Accepted: 02/07/2018] [Indexed: 12/29/2022]
Abstract
Endogenous circadian rhythms are thought to modulate responses to external factors, but mechanisms that confer time-of-day differences in organismal responses to environmental insults/therapeutic treatments are poorly understood. Using a xenobiotic, we find that permeability of the Drosophila "blood"-brain barrier (BBB) is higher at night. The permeability rhythm is driven by circadian regulation of efflux and depends on a molecular clock in the perineurial glia of the BBB, although efflux transporters are restricted to subperineurial glia (SPG). We show that transmission of circadian signals across the layers requires cyclically expressed gap junctions. Specifically, during nighttime, gap junctions reduce intracellular magnesium ([Mg2+]i), a positive regulator of efflux, in SPG. Consistent with lower nighttime efflux, nighttime administration of the anti-epileptic phenytoin is more effective at treating a Drosophila seizure model. These findings identify a novel mechanism of circadian regulation and have therapeutic implications for drugs targeted to the central nervous system.
Collapse
Affiliation(s)
- Shirley L Zhang
- Center for Sleep and Circadian Neurobiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Chronobiology Program at Penn and Howard Hughes Medical Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zhifeng Yue
- Chronobiology Program at Penn and Howard Hughes Medical Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Denice M Arnold
- Chronobiology Program at Penn and Howard Hughes Medical Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gregory Artiushin
- Chronobiology Program at Penn and Howard Hughes Medical Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Amita Sehgal
- Center for Sleep and Circadian Neurobiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Chronobiology Program at Penn and Howard Hughes Medical Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
15
|
Qiankun L, Lanfang M, Xiaojuan D, Yunxia L, Yuan Y, Jingjing L, Junhong L, Longde W, Hongfang L. Pingwei capsules improve gastrointestinal motility in rats with functional dyspepsia. J TRADIT CHIN MED 2018. [DOI: 10.1016/j.jtcm.2018.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
16
|
Liang Q, Yan Y, Mao L, Du X, Liang J, Liu J, Wang L, Li H. Evaluation of a modified rat model for functional dyspepsia. Saudi J Gastroenterol 2018; 24:228-235. [PMID: 29652029 PMCID: PMC6080150 DOI: 10.4103/sjg.sjg_505_17] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND/AIM The purpose of this study was to establish a modified rat model with functional dyspepsia (FD) and analyze the changes in gastrointestinal motility and brain-gut peptide levels in serum and brain-gut axis. MATERIALS AND METHODS Male Wistar rats were divided into control group (Con) and FD model group. FD model was established by stimulating semi-starvation rats via tail damping, provocation, and forced exercise fatigue until gastrointestinal motility disorder appeared, and then levels of motilin, leptin, cholecystokinin (CCK), and vasoactive intestinal peptide (VIP) were detected in serum by enzyme linked immunosorbent assay and in duodenum, antrum, and hypothalamus by immunohistochemistry, reverse transcriptase-polymerase chain reaction, and Western blot. RESULTS The results showed rates of intestinal propulsion and gastric emptying slowed down markedly compared to Con (P < 0.05), the gastrointestinal electric activity attenuated, and migrating motor complex (MMC) interrupted in the model group. The levels of leptin and VIP markedly increased, but motilin decreased as compared to the Con (P < 0.05) in serum and in the above tissues. It is interesting that the level of CCK decreased in the antrum and duodenum but increased in the hypothalamus as compared to Con (P < 0.05). CONCLUSIONS The modified rat model meets the diagnostic criteria of FD and can be used as a method for studying FD in animals.
Collapse
Affiliation(s)
- Qiankun Liang
- Department of Physiology, College of Basic Medicine, Lanzhou University, Lanzhou, China
| | - Yuan Yan
- Department of Physiology, College of Basic Medicine, Lanzhou University, Lanzhou, China
| | - Lanfang Mao
- Department of Gastroenterology, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| | - Xiaojuan Du
- Department of Gastroenterology, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| | - Jingjing Liang
- Department of Physiology, College of Basic Medicine, Lanzhou University, Lanzhou, China
| | - Junhong Liu
- Department of Gastroenterology, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| | - Longde Wang
- Department of Gastroenterology, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| | - Hongfang Li
- Department of Physiology, College of Basic Medicine, Lanzhou University, Lanzhou, China,Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou, China,Address for correspondence: Prof. Hongfang Li, Department of Physiology, College of Basic Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou - 730 000, People's Republic of China. E-mail:
| |
Collapse
|
17
|
Landgraf D, Neumann AM, Oster H. Circadian clock-gastrointestinal peptide interaction in peripheral tissues and the brain. Best Pract Res Clin Endocrinol Metab 2017; 31:561-571. [PMID: 29224668 DOI: 10.1016/j.beem.2017.10.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Food intake and sleep are two mutually exclusive behaviors and both are normally confined to opposing phases of the diurnal cycle. The temporal coordination of behavior and physiology along the 24-h day-night cycle is organized by a network of circadian clocks that orchestrate transcriptional programs controlling cellular physiology. Many of the peptide hormones of the gastrointestinal tract are not only secreted in a circadian fashion, they can also affect circadian clock function in peripheral metabolic tissues and the brain, thus providing metabolic feedback to metabolic and neurobehavioral circuits. In this review, we summarize the current knowledge on this gastrointestinal peptide crosstalk and its potential role in the coordination of nutrition and the maintenance of metabolic homeostasis.
Collapse
Affiliation(s)
- Dominic Landgraf
- Department of Psychiatry, Ludwig Maximilian University of Munich, Germany
| | - Anne-Marie Neumann
- Institute of Neurobiology, Center of Brain, Behavior & Metabolism, University of Lübeck, Germany
| | - Henrik Oster
- Institute of Neurobiology, Center of Brain, Behavior & Metabolism, University of Lübeck, Germany.
| |
Collapse
|
18
|
Association of maternal breast milk and serum levels of macronutrients, hormones, and maternal body composition with infant's body weight. Eur J Clin Nutr 2017; 72:394-400. [PMID: 29167577 DOI: 10.1038/s41430-017-0022-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Revised: 07/05/2017] [Accepted: 09/15/2017] [Indexed: 12/18/2022]
Abstract
BACKGROUND/OBJECTIVES This study was aimed to investigate the association of maternal serum and breast-milk levels of macronutrients, hormones, growth factors, and maternal body composition with infant's body weight. SUBJECTS/METHODS Eighty mother-infant pairs comprised 40 with overweight or obese infant and 40 with normal-weight infant were enrolled in this study. The level of ghrelin, Leptin, adiponectin, EGF, and IGF1 in plasma and breast milk were assessed. Daily breast milk intake and macronutrient concentration along with anthropometric indices of mother-infant pairs were also assessed. RESULTS No significant differences were observed in concentrations of serum hormones between two groups (p > 0.05). However, hormones levels in maternal serum were higher than those in breast milk. A significant positive correlation was found between serum EGF and ghrelin (r = 0.57, p = 0 < 0001). Higher IGF1 in serum showed a significant association with its milk counterpart (r = 0.37). Current mother's weight was associated with infant's weight at the 2nd and 6th month (B = 0.023 p = 0.04, B = 0.055 p = 0.005). The breast-milk macronutrient content was not comparable between two groups. However, the average daily breast milk consumption in obese infants was higher than normals (p = 0.001). Milk EGF and leptin were related to a decrease of 59% and 46% the odds of obese infant development, respectively. There was a significant association of milk EGF and ghrelin with birth weight (B = -0.19, p = 0.04 and B = -0.2, p = 0.04, respectively), and also serum leptin with infant's body weight at the 6th month. CONCLUSIONS Our findings provide a positive association of maternal weight, daily breast milk intake, EGF, and ghrelin with infant's body weight.
Collapse
|
19
|
Abstract
Sleep and its disorders are known to affect the functions of essential organs and systems in the body. However, very little is known about how the blood-brain barrier (BBB) is regulated. A few years ago, we launched a project to determine the impact of sleep fragmentation and chronic sleep restriction on BBB functions, including permeability to fluorescent tracers, tight junction protein expression and distribution, glucose and other solute transporter activities, and mediation of cellular mechanisms. Recent publications and relevant literature allow us to summarize here the sleep-BBB interactions in five sections: (1) the structural basis enabling the BBB to serve as a huge regulatory interface; (2) BBB transport and permeation of substances participating in sleep-wake regulation; (3) the circadian rhythm of BBB function; (4) the effect of experimental sleep disruption maneuvers on BBB activities, including regional heterogeneity, possible threshold effect, and reversibility; and (5) implications of sleep disruption-induced BBB dysfunction in neurodegeneration and CNS autoimmune diseases. After reading the review, the general audience should be convinced that the BBB is an important mediating interface for sleep-wake regulation and a crucial relay station of mind-body crosstalk. The pharmaceutical industry should take into consideration that sleep disruption alters the pharmacokinetics of BBB permeation and CNS drug delivery, being attentive to the chrono timing and activation of co-transporters in subjects with sleep disorders.
Collapse
Affiliation(s)
- Weihong Pan
- 1 Biopotentials Sleep Center, Baton Rouge, LA 70809
| | - Abba J Kastin
- 2 Blood-Brain Barrier Group, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| |
Collapse
|
20
|
Gonzalez-Carter D, Goode AE, Fiammengo R, Dunlop IE, Dexter DT, Porter AE. Inhibition of Leptin-ObR Interaction Does not Prevent Leptin Translocation Across a Human Blood-Brain Barrier Model. J Neuroendocrinol 2016; 28. [PMID: 27037668 DOI: 10.1111/jne.12392] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 03/31/2016] [Accepted: 03/31/2016] [Indexed: 01/01/2023]
Abstract
The adipocyte-derived hormone leptin regulates appetite and energy homeostasis through the activation of leptin receptors (ObR) on hypothalamic neurones; hence, leptin must be transported through the blood-brain barrier (BBB) to reach its target sites in the central nervous system. During obesity, however, leptin BBB transport is decreased, in part precluding leptin as a viable clinical therapy against obesity. Although the short isoform of the ObR (ObRa) has been implicated in the transport of leptin across the BBB as a result of its elevated expression in cerebral microvessels, accumulating evidence indicates that leptin BBB transport is independent of ObRa. In the present study, we employed an ObR-neutralising antibody (9F8) to directly examine the involvement of endothelial ObR in leptin transport across an in vitro human BBB model composed of the human endothelial cell line hCMEC/D3. Our results indicate that, although leptin transport across the endothelial monolayer was nonparacellular, and energy- and endocytosis-dependent, it was not inhibited by pre-treatment with 9F8, despite the ability of the latter to recognise hCMEC/D3-expressed ObR, prevent leptin-ObR binding and inhibit leptin-induced signal transducer and activator of transcription 3 (STAT-3) phosphorylation in hCMEC/D3 cells. Furthermore, hCMEC/D3 cells expressed the transporter protein low-density lipoprotein receptor-related protein-2 (LRP-2), which is capable of binding and endocytosing leptin. In conclusion, our results demonstrate that leptin binding to and signalling through ObR is not required for efficient transport across human endothelial monolayers, indicating that ObR is not the primary leptin transporter at the human BBB, a role which may fall upon LRP-2. A deeper understanding of leptin BBB transport will help clarify the exact causes for leptin resistance seen in obesity and aid in the development of more efficient BBB-penetrating leptin analogues.
Collapse
Affiliation(s)
- D Gonzalez-Carter
- Centre for Neuroinflammation and Neurodegeneration, Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK
- Department of Materials, Faculty of Engineering, Imperial College London, London, UK
| | - A E Goode
- Department of Materials, Faculty of Engineering, Imperial College London, London, UK
| | - R Fiammengo
- Centre for Biomolecular Nanotechnologies @ UniLe, Istituto Italiano di Tecnologia (ITT), Arnesano, Lecce, Italy
| | - I E Dunlop
- Department of Materials, Faculty of Engineering, Imperial College London, London, UK
| | - D T Dexter
- Centre for Neuroinflammation and Neurodegeneration, Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK
| | - A E Porter
- Department of Materials, Faculty of Engineering, Imperial College London, London, UK
| |
Collapse
|
21
|
Pan W. From blood to brain through BBB and astrocytic signaling. Peptides 2015; 72:121-7. [PMID: 26111490 DOI: 10.1016/j.peptides.2015.06.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 06/11/2015] [Accepted: 06/12/2015] [Indexed: 12/14/2022]
Abstract
In this Festschrift, I discuss the career and guiding principles to which Abba J. Kastin has adhered during the last 20 years we worked together. I briefly describe the history of our joint laboratory group, the context of studies of peptide permeation across the blood-brain barrier (BBB), and newer developments in the BBB Group as Abba steps down after serving 35 years as the founding Editor-in-Chief for Peptides. Abba's BBB studies on peptides have contributed to concepts in the neuroendocrinology of feeding and developed information on molecular trafficking across BBB cells. The astroglial leptin signaling studies and the interactions of sleep and BBB are two major directions, whereas the long-term MIF-1 project demarcates a tortuous road on translational research.
Collapse
Affiliation(s)
- Weihong Pan
- Biopotentials Sleep Center, Baton Rouge, LA 70809, USA.
| |
Collapse
|
22
|
Balland E, Cowley MA. New insights in leptin resistance mechanisms in mice. Front Neuroendocrinol 2015; 39:59-65. [PMID: 26410445 DOI: 10.1016/j.yfrne.2015.09.004] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 09/22/2015] [Accepted: 09/23/2015] [Indexed: 11/18/2022]
Abstract
Leptin resistance is one of the main challenges of obesity. To date, two levels of resistance have been identified, first a decreased rate of leptin uptake into the brain and secondly a diminished central response to leptin. New findings have identified the mechanisms of leptin transport and demonstrated that it can be rescued in obesity, but it did not overcome the problem of central resistance. Alteration in the actions of leptin following diet-induced obesity (DIO) appears to be a multifactorial condition. Several phosphatases are inhibiting leptin signaling pathways in a pathological way. Besides, hypothalamic inflammation alters the neuronal circuits that control metabolism. Recent studies describing both mechanisms (inhibition of leptin signaling and inflammation), have provided key insights to potential new targets for treatment. However, recent data showing that DIO mice may conserve a cellular and physiological response to endogenous leptin, highlights the need to redefine the concept of "leptin resistance".
Collapse
Affiliation(s)
- Eglantine Balland
- Department of Physiology, Monash Obesity and Diabetes Institute, Monash University, Clayton, VIC 3800, Australia.
| | - Michael A Cowley
- Department of Physiology, Monash Obesity and Diabetes Institute, Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|
23
|
Page-Wilson G, Meece K, White A, Rosenbaum M, Leibel RL, Smiley R, Wardlaw SL. Proopiomelanocortin, agouti-related protein, and leptin in human cerebrospinal fluid: correlations with body weight and adiposity. Am J Physiol Endocrinol Metab 2015; 309:E458-65. [PMID: 26152765 PMCID: PMC4556883 DOI: 10.1152/ajpendo.00206.2015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 07/01/2015] [Indexed: 11/22/2022]
Abstract
Leptin and its neuronal targets, which produce proopiomelanocortin (POMC) and agouti-related protein (AgRP), regulate energy balance. This study characterized leptin, POMC, and AgRP in the cerebrospinal fluid (CSF) of 47 healthy human subjects, 23 lean and 24 overweight/obese (OW/OB), as related to BMI, adiposity, plasma leptin, soluble leptin receptor (s-OB-R), and insulin. POMC was measured since the POMC prohormone is the predominant POMC peptide in CSF and correlates with hypothalamic POMC in rodents. Plasma AgRP was similarly characterized. CSF leptin was 83-fold lower than in plasma and correlated strongly with BMI, body fat, and insulin. The relative amount of leptin transported into CSF declined with increasing BMI, ranging from 4.5 to 0.52%, consistent with a saturable transport mechanism. CSF sOB-R was 78-fold lower than in plasma and correlated negatively with plasma and CSF leptin. CSF POMC was higher in lean vs. OW/OB subjects (P < 0.001) and correlated negatively with CSF leptin (r = -0.60, P < 0.001) and with plasma leptin, insulin, BMI, and adiposity. CSF AgRP was not different in lean vs. OW/OB; however, plasma AgRP was higher in lean subjects (P = 0.001) and correlated negatively with BMI, adiposity, leptin, insulin, and HOMA (P < 0.005). Thus, CSF measurements may provide useful biomarkers for brain leptin and POMC activity. The striking negative correlation between CSF leptin and POMC could be secondary to leptin resistance and/or neuronal changes associated with obesity but may also indicate that POMC plays a primary role in regulating body weight and adiposity. The role of plasma AgRP as a neuroendocrine biomarker deserves further study.
Collapse
Affiliation(s)
- Gabrielle Page-Wilson
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York
| | - Kana Meece
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York
| | - Anne White
- Faculties of Life Sciences and Medical and Human Sciences, University of Manchester, Manchester, United Kingdom
| | - Michael Rosenbaum
- Department of Pediatrics, Columbia University College of Physicians and Surgeons, New York, New York; and
| | - Rudolph L Leibel
- Department of Pediatrics, Columbia University College of Physicians and Surgeons, New York, New York; and
| | - Richard Smiley
- Department of Anesthesiology, Columbia University College of Physicians and Surgeons, New York, New York
| | - Sharon L Wardlaw
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York;
| |
Collapse
|
24
|
Abstract
The blood-brain barrier (BBB) is a large regulatory and exchange interface between the brain and peripheral circulation. We propose that changes of the BBB contribute to many pathophysiological processes in the brain of subjects with chronic sleep restriction (CSR). To achieve CSR that mimics a common pattern of human sleep loss, we quantified a new procedure of sleep disruption in mice by a week of consecutive sleep recording. We then tested the hypothesis that CSR compromises microvascular function. CSR not only diminished endothelial and inducible nitric oxide synthase, endothelin1, and glucose transporter expression in cerebral microvessels of the BBB, but it also decreased 2-deoxy-glucose uptake by the brain. The expression of several tight junction proteins also was decreased, whereas the level of cyclooxygenase-2 increased. This coincided with an increase of paracellular permeability of the BBB to the small tracers sodium fluorescein and biotin. CSR for 6 d was sufficient to impair BBB structure and function, although the increase of paracellular permeability returned to baseline after 24 h of recovery sleep. This merits attention not only in neuroscience research but also in public health policy and clinical practice.
Collapse
|
25
|
Arnoldussen IAC, Kiliaan AJ, Gustafson DR. Obesity and dementia: adipokines interact with the brain. Eur Neuropsychopharmacol 2014; 24:1982-99. [PMID: 24704273 PMCID: PMC4169761 DOI: 10.1016/j.euroneuro.2014.03.002] [Citation(s) in RCA: 159] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 03/11/2014] [Indexed: 12/20/2022]
Abstract
Obesity is a pandemic and a serious global health concern. Obesity is a risk factor for multiple conditions and contributes to multi-morbidities, resulting in increased health costs and millions of deaths each year. Obesity has been associated with changes in brain structure, cognitive deficits, dementia and Alzheimer׳s disease. Adipokines, defined as hormones, cytokines and peptides secreted by adipose tissue, may have more widespread influence and functionality in the brain than previously thought. In this review, six adipokines, and their actions in the obese and non-obese conditions will be discussed. Included are: plasminogen activator inhibitor-1 (PAI-1), interleukin-6 (IL-6), tumor necrosis factors alpha (TNF-α), angiotensinogen (AGT), adiponectin and leptin. Their functionality in the periphery, their ability to cross the blood brain barrier (BBB) and their influence on dementia processes within the brain will be discussed.
Collapse
Affiliation(s)
- Ilse A C Arnoldussen
- Department of Anatomy, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, Geert Grooteplein Noord 21, 6525 EZ Nijmegen, The Netherlands.
| | - Amanda J Kiliaan
- Department of Anatomy, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, Geert Grooteplein Noord 21, 6525 EZ Nijmegen, The Netherlands.
| | - Deborah R Gustafson
- Department of Neurology, State University of New York-Downstate Medical Center, 450 Clarkson Avenue, Box 1213, Brooklyn, NY11203, USA; UMS 011 Inserm Versailles Saint Quentin, France; Section for Psychiatry and Neurochemistry, Neuropsychiatric Epidemiology Unit, Sahlgrenska Academy at University of Gothenburg, Institute for Neuroscience and Physiology, NeuroPsychiatric Epidemiology Unit, Wallinsgatan 6, 431 41 Gothenburg, Sweden.
| |
Collapse
|
26
|
Ouyang S, Hsuchou H, Kastin AJ, Mishra PK, Wang Y, Pan W. Leukocyte infiltration into spinal cord of EAE mice is attenuated by removal of endothelial leptin signaling. Brain Behav Immun 2014; 40:61-73. [PMID: 24576482 PMCID: PMC4131983 DOI: 10.1016/j.bbi.2014.02.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 02/04/2014] [Accepted: 02/06/2014] [Indexed: 01/17/2023] Open
Abstract
Leptin, a pleiotropic adipokine, crosses the blood-brain barrier (BBB) and blood-spinal cord barrier (BSCB) from the periphery and facilitates experimental autoimmune encephalomyelitis (EAE). EAE induces dynamic changes of leptin receptors in enriched brain and spinal cord microvessels, leading to further questions about the potential roles of endothelial leptin signaling in EAE progression. In endothelial leptin receptor specific knockout (ELKO) mice, there were lower EAE behavioral scores in the early phase of the disorder, better preserved BSCB function shown by reduced uptake of sodium fluorescein and leukocyte infiltration into the spinal cord. Flow cytometry showed that the ELKO mutation decreased the number of CD3 and CD45 cells in the spinal cord, although immune cell profiles in peripheral organs were unchanged. Not only were CD4(+) and CD8(+) T lymphocytes reduced, there were also lower numbers of CD11b(+)Gr1(+) granulocytes in the spinal cord of ELKO mice. In enriched microvessels from the spinal cord of the ELKO mice, the decreased expression of mRNAs for a few tight junction proteins was less pronounced in ELKO than WT mice, as was the elevation of mRNA for CCL5, CXCL9, IFN-γ, and TNF-α. Altogether, ELKO mice show reduced inflammation at the level of the BSCB, less leukocyte infiltration, and better preserved tight junction protein expression and BBB function than WT mice after EAE. Although leptin concentrations were high in ELKO mice and microvascular leptin receptors show an initial elevation before inhibition during the course of EAE, removal of leptin signaling helped to reduce disease burden. We conclude that endothelial leptin signaling exacerbates BBB dysfunction to worsen EAE.
Collapse
Affiliation(s)
- Suidong Ouyang
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | - Hung Hsuchou
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | - Abba J Kastin
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | - Pramod K Mishra
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | - Yuping Wang
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | - Weihong Pan
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA.
| |
Collapse
|
27
|
Wardlaw SL, Burant CF, Klein S, Meece K, White A, Kasten T, Lucey BP, Bateman RJ. Continuous 24-hour leptin, proopiomelanocortin, and amino acid measurements in human cerebrospinal fluid: correlations with plasma leptin, soluble leptin receptor, and amino acid levels. J Clin Endocrinol Metab 2014; 99:2540-8. [PMID: 24670082 PMCID: PMC4079306 DOI: 10.1210/jc.2013-4087] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
CONTEXT In order to characterize diurnal changes in central leptin and its target neuropeptide, proopiomelanocortin (POMC), we measured leptin and POMC in cerebrospinal fluid (CSF) as related to changes in plasma leptin and soluble leptin receptor (sOB-R) levels. CSF and plasma levels of 20 amino acids (AA) were also measured because AA can affect brain POMC. DESIGN AND PARTICIPANTS Stored CSF and plasma samples obtained from eight healthy subjects who served as controls for a previous study were evaluated. CSF was collected hourly over 33 h via indwelling subarachnoid catheter. Leptin, sOB-R, and POMC were measured by sensitive ELISA and AA by gas chromatography-mass spectrometry. RESULTS There was a diurnal rhythm for plasma leptin with a peak at 2200 h (144% of baseline) and there was a similar diurnal rhythm for CSF leptin with a peak (117%) 3-5 h after the plasma peak. Plasma sOB-R was lowest at 0300 h and correlated negatively with plasma and CSF leptin. A diurnal rhythm for POMC in CSF was also detected with a peak (125%) at 0100 h. A positive correlation existed between CSF POMC and leptin in individual subjects over time. CSF levels of many AA increased at night. There was a significant correlation between CSF POMC and 10 AA, including leucine, isoleucine, tryptophan, and tyrosine. CONCLUSIONS Diurnal changes occur in leptin and POMC in human CSF that likely reflect changes in central leptin and melanocortin activity. Our results suggest that nocturnal elevations in leptin, AA, and POMC may help to suppress appetite and feeding at night.
Collapse
Affiliation(s)
- Sharon L Wardlaw
- Department of Medicine (S.L.W., K.M.), Columbia University College of Physicians & Surgeons, New York, New York 10032; Department of Internal Medicine (C.F.B.), University of Michigan Medical School, Ann Arbor, Michigan 48019; Center for Human Nutrition and Atkins Center for Excellence in Obesity Medicine (S.K.), Washington University School of Medicine, St Louis, Missouri 63110; Faculties of Life Sciences and Medical and Human Sciences (A.W.), University of Manchester, Manchester M13 9PL, United Kingdom; and Department of Neurology (T.K., B.P.L., R.J.B), Washington University School of Medicine, St Louis, Missouri 63110
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Pan W, Kastin AJ. Leptin: a biomarker for sleep disorders? Sleep Med Rev 2013; 18:283-90. [PMID: 24080454 DOI: 10.1016/j.smrv.2013.07.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Revised: 06/05/2013] [Accepted: 07/18/2013] [Indexed: 12/11/2022]
Abstract
Leptin, a pleiotropic protein hormone produced mainly by fat cells, regulates metabolic activity and many other physiological functions. The intrinsic circadian rhythm of blood leptin is modulated by gender, development, feeding, fasting, sleep, obesity, and endocrine disorders. Hyperleptinemia is implicated in leptin resistance. To determine the specificity and sensitivity of leptin concentrations in sleep disorders, we summarize here the alterations of leptin in four conditions in animal and human studies: short duration of sleep, sleep fragmentation, obstructive sleep apnea (OSA), and after use of continuous positive airway pressure (CPAP) to treat OSA. The presence and causes of contradictory findings are discussed. Though sustained insufficient sleep lowers fasting blood leptin and therefore probably contributes to increased appetite, obesity and OSA independently result in hyperleptinemia. Successful treatment of OSA by CPAP is predicted to decrease hyperleptinemia, making leptin an ancillary biomarker for treatment efficacy. Current controversies also call for translational studies to determine how sleep disorders regulate leptin homeostasis and how the information can be used to improve sleep treatment.
Collapse
Affiliation(s)
- Weihong Pan
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA 70808, USA.
| | - Abba J Kastin
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA 70808, USA
| |
Collapse
|
29
|
Hypersomnolence and reduced activity in pan-leptin receptor knockout mice. J Mol Neurosci 2013; 51:1038-45. [PMID: 23955775 DOI: 10.1007/s12031-013-0093-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 08/06/2013] [Indexed: 12/26/2022]
Abstract
Excessive obesity correlates with hypersomnolence and impaired cognitive function, presumably induced by metabolic factors and cytokines. Production of the adipokine leptin correlates with the amount of adiposity, and leptin has been shown to promote sleep. To determine whether leptin plays a major role in the hypersomnolence of obesity, we measured sleep architecture in pan-leptin receptor knockout (POKO) mice that do not respond to leptin because of the production of a mutant, non-signaling receptor. The obese POKO mice had more non-rapid eye movement (NREM) sleep and less waking time than their littermate controls. This was mainly seen during the light span, although increased bouts of rapid eye movement sleep were also seen in the dark span. The increase of NREM sleep correlated with the extent of obesity. The POKO mice also had decreased locomotor activity and more immobility in the open field test, but there was no increase of forced immobility nor reduction of sucrose intake as would be seen in depression. The increased NREM sleep and reduced locomotor activity in the POKO mice suggest that it was obesity, rather than leptin signaling, that played a predominant role in altering sleep architecture and activity.
Collapse
|
30
|
Hsuchou H, Wang Y, Cornelissen-Guillaume GG, Kastin AJ, Jang E, Halberg F, Pan W. Diminished leptin signaling can alter circadian rhythm of metabolic activity and feeding. J Appl Physiol (1985) 2013; 115:995-1003. [PMID: 23869060 DOI: 10.1152/japplphysiol.00630.2013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Leptin, a hormone mainly produced by fat cells, shows cell-specific effects to regulate feeding and metabolic activities. We propose that an important feature of metabolic dysregulation resulting in obesity is the loss of the circadian rhythm of biopotentials. This was tested in the pan-leptin receptor knockout (POKO) mice newly generated in our laboratory. In the POKO mice, leptin no longer induced pSTAT-3 signaling after intracerebroventricular injection. Three basic phenotypes were observed: the heterozygotes had similar weight and adiposity as the wild-type (WT) mice (>60% of the mice); the homozygotes were either fatter (∼30%), or rarely leaner (<5%) than the WT mice. By early adulthood, the POKO mice had higher average body weight and adiposity than their respective same-sex WT littermate controls, and this was consistent among different batches. The homozygote fat POKO showed significant reduction of midline estimating statistic of rhythm of circadian parameters, and shifts of ultradian rhythms. The blunted circadian rhythm of these extremely obese POKO mice was also seen in their physical inactivity, longer feeding bouts, and higher food intake. The extent of obesity correlated with the blunted circadian amplitude, accumulative metabolic and locomotor activities, and the severity of hyperphagia. This contrasts with the heterozygote POKO mice which showed little obesity and metabolic disturbance, and only subtle changes of the circadian rhythm of metabolic activity without alterations in feeding behavior. The results provide a novel aspect of leptin resistance, almost manifesting as an "all or none" phenomenon.
Collapse
Affiliation(s)
- Hung Hsuchou
- Blood-Brain Barrier Group, Pennington Biomedical Research Center, Baton Rouge, Louisiana
| | | | | | | | | | | | | |
Collapse
|
31
|
Pan W, Stone KP, Hsuchou H, Manda VK, Zhang Y, Kastin AJ. Cytokine signaling modulates blood-brain barrier function. Curr Pharm Des 2013; 17:3729-40. [PMID: 21834767 DOI: 10.2174/138161211798220918] [Citation(s) in RCA: 164] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 08/30/2011] [Accepted: 09/26/2011] [Indexed: 02/06/2023]
Abstract
The blood-brain barrier (BBB) provides a vast interface for cytokines to affect CNS function. The BBB is a target for therapeutic intervention. It is essential, therefore, to understand how cytokines interact with each other at the level of the BBB and how secondary signals modulate CNS functions beyond the BBB. The interactions between cytokines and lipids, however, have not been fully addressed at the level of the BBB. Here, we summarize current understanding of the localization of cytokine receptors and transporters in specific membrane microdomains, particularly lipid rafts, on the luminal (apical) surface of the microvascular endothelial cells composing the BBB. We then illustrate the clinical context of cytokine effects on the BBB by neuroendocrine regulation and amplification of inflammatory signals. Two unusual aspects discussed are signaling crosstalk by different classes of cytokines and genetic regulation of drug efflux transporters. We also introduce a novel area of focus on how cytokines may act through nuclear hormone receptors to modulate efflux transporters and other targets. A specific example discussed is the ATP-binding cassette transporter-1 (ABCA-1) that regulates lipid metabolism. Overall, cytokine signaling at the level of the BBB is a crucial feature of the dynamic regulation that can rapidly change BBB function and affect brain health and disease.
Collapse
Affiliation(s)
- Weihong Pan
- Blood-Brain Barrier Group, Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA 70808, USA
| | | | | | | | | | | |
Collapse
|
32
|
Ishihara Y, White CL, Kageyama H, Kageyama A, York DA, Bray GA. Effects of Diet and Time of the Day on Serum and CSF Leptin Levels in Osborne-Mendel and S5B/Pl Rats. ACTA ACUST UNITED AC 2012; 12:1067-76. [PMID: 15292470 DOI: 10.1038/oby.2004.134] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVE To characterize the effects of dietary fat on the diurnal variation in serum and cerebrospinal fluid (CSF) leptin levels in Osborne-Mendel (OM) and S5B/Pl rats and quantitate the dose response to lower doses of leptin administered into the third cerebral ventricle. RESEARCH METHODS AND PROCEDURES Rats were fitted with implanted vascular ports or third ventricular cannulas and fed either laboratory chow or one of two semipurified high-fat or low-fat diets. Leptin and insulin were measured by immunoassay. RESULTS Serum leptin and insulin levels were positively correlated and had similar patterns of diurnal change. CSF leptin and insulin also had diurnal rhythms, with a peak at 7:00 am, but the diurnal oscillations of leptin and insulin were significantly lower in the S5B/Pl rats than the OM rats. Thus, the ratio of CSF to serum leptin was significantly higher in the S5B/Pl rats than in the OM rats. Dietary fat had no effect on these diurnal patterns. There was a right shift in the dose response to leptin in the OM rats compared with the S5B/P1 rats. S5B/P1 rats treated with leptin had higher signal transduction and translation (STAT-3) mRNA levels compared with pair-fed or saline injected S5B/P1 rats. Hypothalamic suppressors of cytokine signaling mRNA levels were not statistically different between the groups. DISCUSSION The higher CSF-to-serum leptin ratio in the S5B/P1 rats, the enhanced suppression of food intake and body weight with leptin injections, and the higher STAT-3 activity in these animals suggest that S5B/P1 rats are more sensitive to leptin than OM rats.
Collapse
Affiliation(s)
- Yuri Ishihara
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge 70808, USA
| | | | | | | | | | | |
Collapse
|
33
|
Abstract
The blood-brain barrier (BBB) regulates the blood-to-brain passage of gastrointestinal hormones, thus informing the brain about feeding and nutritional status. Disruption of this communication results in dysregulation of feeding and body weight control. Leptin, which crosses the BBB to inform the CNS about adiposity, provides an example. Impaired leptin transport, especially coupled with central resistance, results in obesity. Various substances/conditions regulate leptin BBB transport. For example, triglycerides inhibit leptin transport. This may represent an evolutionary adaptation in that hypertriglyceridemia occurs during starvation. Inhibition of leptin, an anorectic, during starvation could have survival advantages. The large number of other substances that influence feeding is explained by the complexity of feeding. This complexity includes cognitive aspects; animals in the wild are faced with cost/benefit analyses to feed in the safest, most economical way. This cognitive aspect partially explains why so many feeding substances affect neurogenesis, neuroprotection, and cognition. The relation between triglycerides and cognition may be partially mediated through triglyceride's ability to regulate the BBB transport of cognitively active gastrointestinal hormones such as leptin, insulin, and ghrelin.
Collapse
Affiliation(s)
- William A Banks
- GRECC, Veterans Affairs Puget Sound Health Care System and Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington 98108, USA.
| |
Collapse
|
34
|
Pan W, Hsuchou H, Jayaram B, Khan RS, Huang EYK, Wu X, Chen C, Kastin AJ. Leptin action on nonneuronal cells in the CNS: potential clinical applications. Ann N Y Acad Sci 2012; 1264:64-71. [PMID: 22530983 PMCID: PMC3407332 DOI: 10.1111/j.1749-6632.2012.06472.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Leptin, an adipocyte-derived cytokine, crosses the blood–brain barrier to act on many regions of the central nervous system (CNS). It participates in the regulation of energy balance, inflammatory processes, immune regulation, synaptic formation, memory condensation, and neurotrophic activities. This review focuses on the newly identified actions of leptin on astrocytes. We first summarize the distribution of leptin receptors in the brain, with a focus on the hypothalamus, where the leptin receptor is known to mediate essential feeding suppression activities, and on the hippocampus, where leptin facilitates memory, reduces neurodegeneration, and plays a dual role in seizures. We will then discuss regulation of the nonneuronal leptin system in obesity. Its relationship with neuronal leptin signaling is illustrated by in vitro assays in primary astrocyte culture and by in vivo studies on mice after pretreatment with a glial metabolic inhibitor or after cell-specific deletion of intracellular signaling leptin receptors. Overall, the glial leptin system shows robust regulation and plays an essential role in obesity. Strategies to manipulate this nonneuronal leptin signaling may have major clinical impact.
Collapse
Affiliation(s)
- Weihong Pan
- Blood-Brain Barrier Group, Pennington Biomedical Research Center, Baton Rouge, Lousiana 70808, USA.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Hsuchou H, Kastin AJ, Tu H, Markadakis EN, Stone KP, Wang Y, Heymsfield SB, Chua SS, Obici S, Magrisso IJ, Pan W. Effects of cell-type specific leptin receptor mutation on leptin transport across the BBB. Peptides 2011; 32:1392-9. [PMID: 21616110 PMCID: PMC3137692 DOI: 10.1016/j.peptides.2011.05.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Revised: 05/09/2011] [Accepted: 05/09/2011] [Indexed: 11/19/2022]
Abstract
The functions of leptin receptors (LRs) are cell-type specific. At the blood-brain barrier, LRs mediate leptin transport that is essential for its CNS actions, and both endothelial and astrocytic LRs may be involved. To test this, we generated endothelia specific LR knockout (ELKO) and astrocyte specific LR knockout (ALKO) mice. ELKO mice were derived from a cross of Tie2-cre recombinase mice with LR-floxed mice, whereas ALKO mice were generated by a cross of GFAP-cre with LR-floxed mice, yielding mutant transmembrane LRs without signaling functions in endothelial cells and astrocytes, respectively. The ELKO mutation did not affect leptin half-life in blood or apparent influx rate to the brain and spinal cord, though there was an increase of brain parenchymal uptake of leptin after in situ brain perfusion. Similarly, the ALKO mutation did not affect blood-brain barrier permeation of leptin or its degradation in blood and brain. The results support our observation from cellular studies that membrane-bound truncated LRs are fully efficient in transporting leptin, and that basal levels of astrocytic LRs do not affect leptin transport across the endothelial monolayer. Nonetheless, the absence of leptin signaling at the BBB appears to enhance the availability of leptin to CNS parenchyma. The ELKO and ALKO mice provide new models to determine the dynamic regulation of leptin transport in metabolic and inflammatory disorders where cellular distribution of LRs is shifted.
Collapse
Affiliation(s)
- Hung Hsuchou
- Pennington Biomedical Research Center, Baton Rouge, LA 70808
| | - Abba J. Kastin
- Pennington Biomedical Research Center, Baton Rouge, LA 70808
| | - Hong Tu
- Pennington Biomedical Research Center, Baton Rouge, LA 70808
| | | | | | - Yuping Wang
- Pennington Biomedical Research Center, Baton Rouge, LA 70808
| | | | | | - Silvana Obici
- Obesity Research Center, University of Cincinnati, Cincinnati, Ohio 45237
| | - I. Jack Magrisso
- Obesity Research Center, University of Cincinnati, Cincinnati, Ohio 45237
| | - Weihong Pan
- Pennington Biomedical Research Center, Baton Rouge, LA 70808
| |
Collapse
|
36
|
Ando H, Kumazaki M, Motosugi Y, Ushijima K, Maekawa T, Ishikawa E, Fujimura A. Impairment of peripheral circadian clocks precedes metabolic abnormalities in ob/ob mice. Endocrinology 2011; 152:1347-54. [PMID: 21285316 DOI: 10.1210/en.2010-1068] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Recent studies have demonstrated relationships between the dysfunction of circadian clocks and the development of metabolic abnormalities, but the chicken-and-egg question remains unresolved. To address this issue, we investigated the cause-effect relationship in obese, diabetic ob/ob mice. Compared with control C57BL/6J mice, the daily mRNA expression profiles of the clock and clock-controlled genes Clock, Bmal1, Cry1, Per1, Per2, and Dbp were substantially dampened in the liver and adipose tissue, but not the hypothalamic suprachiasmatic nucleus, of 10-wk-old ob/ob mice. Four-week feeding of a low-calorie diet and administration of leptin over a 7-d period attenuated, to a significant and comparable extent, the observed metabolic abnormalities (obesity, hyperglycemia, hyperinsulinemia, and hypercholesterolemia) in the ob/ob mice. However, only leptin treatment improved the impaired peripheral clocks. In addition, clock function, assessed by measuring levels of Per1, Per2, and Dbp mRNA at around peak times, was also reduced in the peripheral tissues of 3-wk-old ob/ob mice without any overt metabolic abnormalities. Collectively these results indicate that the impairment of peripheral clocks in ob/ob mice does not result from metabolic abnormalities but may instead be at least partially caused by leptin deficiency itself. Further studies are needed to clarify how leptin deficiency affects peripheral clocks.
Collapse
Affiliation(s)
- Hitoshi Ando
- Division of Clinical Pharmacology, Department of Pharmacology, School of Medicine, Jichi Medical University, Tochigi 329-0498, Japan
| | | | | | | | | | | | | |
Collapse
|
37
|
Heldsinger A, Grabauskas G, Song I, Owyang C. Synergistic interaction between leptin and cholecystokinin in the rat nodose ganglia is mediated by PI3K and STAT3 signaling pathways: implications for leptin as a regulator of short term satiety. J Biol Chem 2011; 286:11707-15. [PMID: 21270124 PMCID: PMC3064222 DOI: 10.1074/jbc.m110.198945] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Revised: 01/25/2011] [Indexed: 11/06/2022] Open
Abstract
Research has shown that the synergistic interaction between vagal cholecystokinin-A receptors (CCKARs) and leptin receptors (LRbs) mediates short term satiety. We hypothesize that this synergistic interaction is mediated by cross-talk between signaling cascades used by CCKARs and LRbs, which, in turn, activates closure of K(+) channels, leading to membrane depolarization and neuronal firing. Whole cell patch clamp recordings were performed on isolated rat nodose ganglia neurons. Western immunoblots elucidated the intracellular signaling pathways that modulate leptin/CCK synergism. In addition, STAT3, PI3K, Src, and MAPK genes were silenced by lentiviral infection and transient Lipofectamine transfection of cultured rat nodose ganglia to determine the effect of these molecules on leptin/CCK synergism. Patch clamp studies showed that a combination of leptin and CCK-8 caused a significant increase in membrane input resistance compared with leptin or CCK-8 alone. Silencing the STAT3 gene abolished the synergistic action of leptin/CCK-8 on neuronal firing. Leptin/CCK-8 synergistically stimulated a 7.7-fold increase in phosphorylated STAT3 (pSTAT3), which was inhibited by AG490, C3 transferase, PP2, LY294002, and wortmannin, but not PD98059. Silencing the Src and PI3K genes resulted in a loss of leptin/CCK-stimulated pSTAT3. We conclude that the synergistic interaction between vagal CCKARs and LRbs is mediated by the phosphorylation of STAT3, which, in turn, activates closure of K(+) channels, leading to membrane depolarization and neuronal firing. This involves the interaction between CCK/Src/PI3K cascades and leptin/JAK2/PI3K/STAT3 signaling pathways. Malfunctioning of these signaling molecules may result in eating disorders.
Collapse
Affiliation(s)
- Andrea Heldsinger
- From the Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109
| | - Gintautas Grabauskas
- From the Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109
| | - Il Song
- From the Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109
| | - Chung Owyang
- From the Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109
| |
Collapse
|
38
|
Barnes MJ, Rogers RC, Van Meter MJ, Hermann GE. Co-localization of TRHR1 and LepRb receptors on neurons in the hindbrain of the rat. Brain Res 2010; 1355:70-85. [PMID: 20691166 DOI: 10.1016/j.brainres.2010.07.094] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2010] [Revised: 07/24/2010] [Accepted: 07/27/2010] [Indexed: 02/06/2023]
Abstract
We have reported a highly cooperative interaction between leptin and thyrotropin releasing hormone (TRH) in the hindbrain to generate thermogenic responses (Hermann et al., 2006) (Rogers et al., 2009). Identifying the locus in the hindbrain where leptin and TRH act synergistically to increase thermogenesis will be necessary before we can determine the mechanism(s) by which this interaction occurs. Here, we performed heat-induced epitope recovery techniques and in situ hybridization to determine if neurons or afferent fibers in the hindbrain possess both TRH type 1 receptor and long-form leptin receptor [TRHR1; LepRb, respectively]. LepRb receptors were highly expressed in the solitary nucleus [NST], dorsal motor nucleus of the vagus [DMN] and catecholaminergic neurons of the ventrolateral medulla [VLM]. All neurons that contained LepRb also contained TRHR1. Fibers in the NST and the raphe pallidus [RP] and obscurrus [RO] that possess LepRb receptors were phenotypically identified as glutamatergic type 2 fibers (vglut2). Fibers in the NST and RP that possess TRHR1 receptors were phenotypically identified as serotonergic [i.e., immunopositive for the serotonin transporter; SERT]. Co-localization of LepRb and TRHR1 was not observed on individual fibers in the hindbrain but these two fiber types co-mingle in these nuclei. These anatomical arrangements may provide a basis for the synergy between leptin and TRH to increase thermogenesis.
Collapse
Affiliation(s)
- Maria J Barnes
- Laboratory of Autonomic Neuroscience, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA
| | | | | | | |
Collapse
|
39
|
Ziylan YZ, Baltaci AK, Mogulkoc R. Leptin transport in the central nervous system. Cell Biochem Funct 2009; 27:63-70. [PMID: 19205004 DOI: 10.1002/cbf.1538] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Synthesized and released by the adipose tissue, leptin is the widely studied 167-amino acid hormonal protein product of the obesity gene. Originally leptin was defined in association with satiety and energy balance and claimed to be an anti-obesity factor that functioned via a feedback effect from adipocytes to hypothalamus. There is a growing body of evidence that emphasizes the importance of leptin in the regulation of food intake and body weight in animals and humans, alike. Other research findings point out that it plays a role in the regulation of the metabolism, sexual development, reproduction, hematopoiesis, immunity, gastrointestinal functions, sympathetic activation, and angiogenesis. The aim of this review is to evaluate the relation between leptin and the central nervous system (CNS).
Collapse
Affiliation(s)
- Yusuf Ziya Ziylan
- Department of Physiology, Istanbul Medical School, Istanbul University, Capa, Istanbul, Turkey
| | | | | |
Collapse
|
40
|
Raff H. Commentaries on Viewpoint: Effect of altitude on leptin levels, does it go up or down? Control of leptin with altitude exposure. J Appl Physiol (1985) 2008; 105:1686-7. [DOI: 10.1152/japplphysiol.zdg-8233-vpcomm.2008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
41
|
Kastin AJ, Pan W. Blood-brain barrier and feeding: regulatory roles of saturable transport systems for ingestive peptides. Curr Pharm Des 2008; 14:1615-9. [PMID: 18673203 DOI: 10.2174/138161208784705423] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The two main ways for peptides in the peripheral body to enter the brain are by either saturable transport or passive diffusion across the blood-brain barrier (BBB). Saturable transport systems have the advantage of being responsive to physiological and pathological stimuli. Since saturable systems can regulate peptide entry into the brain, they have the potential to play controlling roles in feeding behavior. For therapeutic applications, however, saturable systems have the disadvantage of functioning as a threshold to limit access of large amounts of peptides into the brain. This pharmacological problem presumably would not be encountered for peptides crossing the BBB by passive diffusion, a process dependent on physicochemical properties. Thus, the gatekeeper function of the BBB can be expanded to a primary governing role, especially for entry of ingestive peptides subject to their respective saturable transport systems.
Collapse
Affiliation(s)
- Abba J Kastin
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA.
| | | |
Collapse
|
42
|
Pan W, Hsuchou H, Tu H, Kastin AJ. Developmental changes of leptin receptors in cerebral microvessels: unexpected relation to leptin transport. Endocrinology 2008; 149:877-85. [PMID: 18039787 PMCID: PMC2276815 DOI: 10.1210/en.2007-0893] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The adipokine leptin participates not only in the regulation of feeding and obesity in adults but also in neonatal development. It crosses the blood-brain barrier (BBB) by receptor-mediated transport. Leptin concentrations in blood differ between neonates and adults. We determined the developmental changes of leptin receptor subtypes in the cerebral microvessels composing the BBB and examined their expected correlation with leptin transport across the BBB. Total RNA was extracted from enriched cerebral microvessels of mice 1, 7, 14, and 60 d of age for real-time RT-PCR analysis of leptin receptor subtypes. In cerebral microvessels from neonates, ObRa, ObRb, ObRc, and ObRe mRNA were all higher than in adults, but ObRd was not detectable. Hypothalamus showed similar age-related changes except for ObRb, which was higher in adults. The homologous receptor gp130 did not show significant age-related changes in either region. Despite the increase of leptin receptors, leptin permeation across the BBB after iv injection was less in the neonates. In situ brain perfusion with blood-free buffer showed no significant difference in the brain uptake of leptin between neonates and adults, indicating an antagonistic role of leptin-binding proteins in the circulation, especially the soluble receptor ObRe. The results are consistent with our previous finding that ObRe antagonizes leptin endocytosis in cultured endothelia and transport from blood to brain in mice. Overall, the developmental changes observed for leptin receptors unexpectedly failed to correlate with the entry of leptin into brain, and this may indicate different functions of the receptors in neonates and adults.
Collapse
Affiliation(s)
- Weihong Pan
- Blood-Brain Barrier Group, Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, Louisiana 70808, USA.
| | | | | | | |
Collapse
|
43
|
Sierra-Johnson J, Romero-Corral A, Somers VK, Johnson BD. Effect of altitude on leptin levels, does it go up or down? J Appl Physiol (1985) 2008; 105:1684-5. [PMID: 18276896 DOI: 10.1152/japplphysiol.01284.2007] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Justo Sierra-Johnson
- Division of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | | | | | |
Collapse
|
44
|
Central leptin insufficiency syndrome: an interactive etiology for obesity, metabolic and neural diseases and for designing new therapeutic interventions. Peptides 2007; 29:127-38. [PMID: 18053615 DOI: 10.1016/j.peptides.2007.10.017] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2007] [Revised: 10/17/2007] [Accepted: 10/18/2007] [Indexed: 01/10/2023]
Abstract
This review critically reappraises recent scientific evidence concerning central leptin insufficiency versus leptin resistance formulations to explain metabolic and neural disorders resulting from subnormal or defective leptin signaling in various sites in the brain. Research at various fronts to unravel the complexities of the neurobiology of leptin is surveyed to provide a comprehensive account of the neural and metabolic effects of environmentally imposed fluctuations in leptin availability at brain sites and the outcome of newer technology to restore leptin signaling in a site-specific manner. The cumulative new knowledge favors a unified central leptin insufficiency syndrome over the, in vogue, central resistance hypothesis to explain the global adverse impact of deficient leptin signaling in the brain. Furthermore, the leptin insufficiency syndrome delineates a novel role of leptin in the hypothalamus in restraining rhythmic pancreatic insulin secretion while concomitantly enhancing glucose metabolism and non-shivering thermogenic energy expenditure, sequelae that would otherwise promote fat accrual to store excess energy resulting from consumption of energy-enriched diets. A concerted effort should now focus on development of newer technologies for delivery of leptin or leptin mimetics to specifically target neural pathways for remediation of diverse ailments encompassing the central leptin insufficiency syndrome.
Collapse
|
45
|
Abstract
Just as the blood-brain barrier (BBB) is not a static barrier, the adipocytes are not inert storage depots. Adipokines are peptides or polypeptides produced by white adipose tissue; they play important roles in normal physiology as well as in the metabolic syndrome. Adipokines secreted into the circulation can interact with the BBB and exert potent CNS effects. The specific transport systems for two important adipokines, leptin and tumor necrosis factor alpha, have been characterized during the past decade. By contrast, transforming growth factor beta-1 and adiponectin do not show specific permeation across the BBB, but modulate endothelial functions. Still others, like interleukin-6, may reach the brain but are rapidly degraded. This review summarizes current knowledge and recent findings of the rapidly growing family of adipokines and their interactions with the BBB.
Collapse
Affiliation(s)
- Weihong Pan
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA 70808, United States.
| | | |
Collapse
|
46
|
Aydin S, Aydin S, Ozkan Y, Kumru S. Ghrelin is present in human colostrum, transitional and mature milk. Peptides 2006; 27:878-82. [PMID: 16191452 DOI: 10.1016/j.peptides.2005.08.006] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2005] [Revised: 08/17/2005] [Accepted: 08/17/2005] [Indexed: 01/09/2023]
Abstract
Ghrelin and its mRNA have recently been found in numerous human tissues including breast. The aim of this study was to compare the ghrelin levels in colostrum, mature and transitional milk and plasma in lactating women with plasma samples from non-lactating women. Venous blood samples were obtained from 17 healthy lactating women aged 22-35 years and from 16 age-matched controls. Colostrum, transitional and mature milk samples were collected just before suckling. The level of bioactive ghrelin was determined by RIA. Comparison of ghrelin values for lactating women showed significantly lower concentrations in colostrum (70.3 +/- 18 pg/ml), transitional milk (83.8 +/- 18pg/ml) and mature milk (97.3 +/- 13 pg/ml) than in the corresponding plasma samples (first day 95 +/- 16 pg/ml, 10th day 111 +/- 13 pg/ml and 15th day 135 +/- 16 pg/ml). The plasma concentrations were lower in the lactating than in the non-lactating women. Thus, the ghrelin levels in colostrum, transitional and mature milk were elavated concomitantly with increasing plasma ghrelin after delivery. The origin of milk ghrelin is not known, but it probably comes from the plasma.
Collapse
Affiliation(s)
- Suleyman Aydin
- Department of Biochemistry and Clinical Biochemistry, Firat University, Medical School (Firat Medical Center), 23119 Elazig, Turkey.
| | | | | | | |
Collapse
|
47
|
Wetzler S, Jean-Joseph G, Even P, Tomé D, Larue-Achagiotis C. Acute third ventricular administration of leptin decreases protein and fat in self-selecting rats. Behav Brain Res 2005; 159:119-25. [PMID: 15795005 DOI: 10.1016/j.bbr.2004.10.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2004] [Revised: 10/15/2004] [Accepted: 10/18/2004] [Indexed: 12/24/2022]
Abstract
The peripheral administration of leptin reduces food intake (FI) body weight gain (BWG) and modifies food choice. The aim of this study was to examine the effect of acute cerebral injections of leptin on food selection in rats. Male rats were first adapted to the food choice paradigm (protein, carbohydrate, fat) for 3 weeks. They were then implanted with a cannula in the third ventricle. Leptin (leptin group=L) or saline (control group=C) injections were performed at either the beginning or the end of the night at 4-day intervals. FI was recorded continuously, 3 days before, during and then after injections. Rats were sacrificed 86 h after the second injection. After both injections, BWG and FI were reduced. The reduction in FI concerned only nocturnal intake, whatever the timing of the injection. When the injection was given at the beginning of the night, the reductions after a 1-h latency period were -45% and -27.5% during the first and second days, respectively. Following the second injection, the same effects were observed immediately (-16% and -41%, respectively). Only the fat and protein intakes were significantly reduced. This lower FI was due to a reduction in meal size and duration. The reduction resulted in a lower BWG and total white adipose tissue mass. At the time of sacrifice, 6 h after food deprivation, leptinemia and insulinemia were reduced in leptin-treated rats. Glycemia values were identical. It was thus demonstrated that central leptin was a satiation factor rather than a satiety factor.
Collapse
Affiliation(s)
- Sandrine Wetzler
- UMR INRA/INAPG 914 Physiologie de la Nutrition et du Comportement Alimentaire Institut National Agronomique de Paris-Grignon, 16, rue Claude Bernard, 75231 Paris Cedex 05, France
| | | | | | | | | |
Collapse
|
48
|
Abbott NJ. Physiology of the blood–brain barrier and its consequences for drug transport to the brain. ACTA ACUST UNITED AC 2005. [DOI: 10.1016/j.ics.2005.02.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
49
|
Pan W, Kastin AJ. Why study transport of peptides and proteins at the neurovascular interface. ACTA ACUST UNITED AC 2004; 46:32-43. [PMID: 15297153 DOI: 10.1016/j.brainresrev.2004.04.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2004] [Indexed: 01/17/2023]
Abstract
The blood-brain barrier (BBB) is an immense neurovascular interface. In neurodegenerative, ischemic, and traumatic disorders of the central nervous system (CNS), the BBB may hinder the delivery of many therapeutic peptides and proteins to the brain and spinal cord. Fortunately, the mistaken dogma that peptides and proteins do not cross the BBB has been corrected during the past two decades by the accumulating evidence that peptides and proteins in the periphery exert potent effects in the CNS. Not only can peptides and proteins serve as carriers for selective therapeutic agents, but they themselves may directly cross the BBB after delivery into the bloodstream. Their passage may be mediated by simple diffusion or specific transport, both of which can be affected by interactions in the blood compartment (outside the BBB) and within the endothelial cells (at the BBB level). Although the majority of current delivery strategies focuses on modification of the molecule to be delivered, understanding the mechanisms of transport will eventually facilitate regulation of the BBB directly. We review the different aspects of interactions and discuss recent advances in the cell biology of peptide/protein transport across the BBB. Better understanding of the nature and regulation of the transport systems at the BBB will provide a new direction to enhance the interactions of peripheral peptides and proteins with the CNS.
Collapse
Affiliation(s)
- Weihong Pan
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA 70808, USA.
| | | |
Collapse
|
50
|
Perfetto F, Tarquini R, Cornélissen G, Mello G, Tempestini A, Gaudiano P, Mancuso F, Halberg F. Circadian phase difference of leptin in android versus gynoid obesity. Peptides 2004; 25:1297-306. [PMID: 15350697 DOI: 10.1016/j.peptides.2004.06.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2004] [Revised: 06/02/2004] [Accepted: 06/04/2004] [Indexed: 10/26/2022]
Abstract
A circadian rhythm in serum leptin, measured every 4 h for 24 h, characterizes normal-weight women (N = 14), and women with gynoid (N = 17) or android (N = 26) obesity, peaking around midnight (P < 0.05), but differing by about 3 h between android and gynoid women (P < 0.01). Obesity is associated with a higher MESOR (rhythm-adjusted mean; P < 0.001) and a smaller relative circadian amplitude (P < 0.05). Gynoid obesity is associated with a larger circadian amplitude of cortisol (P < 0.05), whereas android obesity is associated with a larger circadian amplitude and a higher MESOR of insulin (P < 0.05). Understanding putative mechanisms underlying different body fat distribution may lead to improved chronotherapeutic measures.
Collapse
Affiliation(s)
- Federico Perfetto
- Department of Internal Medicine, Facoltà di Medicina e Chirurgia, University of Florence, Viale Pieraccini 18, 50139 Florence, Italy.
| | | | | | | | | | | | | | | |
Collapse
|