51
|
Watts AG, Kanoski SE, Sanchez-Watts G, Langhans W. The physiological control of eating: signals, neurons, and networks. Physiol Rev 2022; 102:689-813. [PMID: 34486393 PMCID: PMC8759974 DOI: 10.1152/physrev.00028.2020] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 08/30/2021] [Indexed: 02/07/2023] Open
Abstract
During the past 30 yr, investigating the physiology of eating behaviors has generated a truly vast literature. This is fueled in part by a dramatic increase in obesity and its comorbidities that has coincided with an ever increasing sophistication of genetically based manipulations. These techniques have produced results with a remarkable degree of cell specificity, particularly at the cell signaling level, and have played a lead role in advancing the field. However, putting these findings into a brain-wide context that connects physiological signals and neurons to behavior and somatic physiology requires a thorough consideration of neuronal connections: a field that has also seen an extraordinary technological revolution. Our goal is to present a comprehensive and balanced assessment of how physiological signals associated with energy homeostasis interact at many brain levels to control eating behaviors. A major theme is that these signals engage sets of interacting neural networks throughout the brain that are defined by specific neural connections. We begin by discussing some fundamental concepts, including ones that still engender vigorous debate, that provide the necessary frameworks for understanding how the brain controls meal initiation and termination. These include key word definitions, ATP availability as the pivotal regulated variable in energy homeostasis, neuropeptide signaling, homeostatic and hedonic eating, and meal structure. Within this context, we discuss network models of how key regions in the endbrain (or telencephalon), hypothalamus, hindbrain, medulla, vagus nerve, and spinal cord work together with the gastrointestinal tract to enable the complex motor events that permit animals to eat in diverse situations.
Collapse
Affiliation(s)
- Alan G Watts
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Scott E Kanoski
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Graciela Sanchez-Watts
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Wolfgang Langhans
- Physiology and Behavior Laboratory, Eidgenössische Technische Hochschule-Zürich, Schwerzenbach, Switzerland
| |
Collapse
|
52
|
Sánchez-Catalán MJ, Barrot M. Fos response of the tail of the ventral tegmental area to food restriction entails a prediction error processing. Behav Brain Res 2022; 425:113826. [PMID: 35247487 DOI: 10.1016/j.bbr.2022.113826] [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: 09/24/2021] [Revised: 02/28/2022] [Accepted: 02/28/2022] [Indexed: 11/25/2022]
Abstract
The tail of the ventral tegmental area (tVTA) or rostromedial tegmental nucleus (RMTg) receives lateral habenula inputs and projects heavily to midbrain dopamine neurons. Midbrain dopamine and lateral habenula neurons participate in learning processes predicting the outcomes of actions, placing the tVTA in a critical location into prediction error pathways. tVTA GABA neurons show electrophysiological inhibition or activation after reward and aversive stimuli, respectively, and their predictive cues. tVTA molecular recruitment, however, is not elicited by all aversive stimuli. Indeed, precipitated opioid withdrawal, repeated footshocks or food restriction raise tVTA Fos expression, whereas various other unpleasant, stressful or painful stimuli does not elicit that molecular response. However, the basis of that difference remains unknown. In the present study, we tried to disentangle whether the tVTA c-Fos induction observed after food restriction was due to the aversive state of food restriction or to procedure-related reward prediction error. To this end, male Sprague-Dawley rats were food-restricted for 7-8 days. During this period, animals were handled and weighed every day before feeding. On the test day, rats underwent several behavioral procedures to explore the impact of food restriction and food-predictive cue exposure on tVTA c-Fos expression. We showed that food restriction per se was not able to recruit c-Fos in the tVTA. On the contrary, the food-predicting cues induced c-Fos locally in the absence of feeding, whereas the food-predicting cues followed by feeding evoked lower c-Fos expression. Overall, our results support the proposed involvement of the tVTA in reward prediction error.
Collapse
Affiliation(s)
- María-José Sánchez-Catalán
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, F-67000 Strasbourg, France; Unitat Predepartamental de Medicina, Universitat Jaume I, Castelló de la Plana, Spain.
| | - Michel Barrot
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, F-67000 Strasbourg, France
| |
Collapse
|
53
|
Brandlhuber M, Benhaqi P, Brandlhuber B, Koliogiannis V, Kasparek MS, Mueller MH, Kreis ME. The role of vagal innervation on the early development of postoperative ileus in mice. Neurogastroenterol Motil 2022; 34:e14308. [PMID: 34962331 DOI: 10.1111/nmo.14308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 05/30/2021] [Accepted: 10/27/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Postoperative ileus (POI) involves an intestinal inflammatory response that is modulated by afferent and efferent vagal activation. We aimed to identify the potential influence of the vagus nerve on POI by tracking central vagal activation and its role for peripheral inflammatory changes during the early hours after surgery. METHODS C57BL6 mice were vagotomized (V) 3-4 days prior to experiments, while control animals received sham vagotomy (SV). Subgroups underwent either laparotomy (sham operation; S-POI) or laparotomy followed by standardized small bowel manipulation to induce postoperative ileus (POI). Three hours and 9 h later, respectively, a jejunal segment was harvested and infiltration of inflammatory cells in intestinal muscularis was evaluated by fluorescein isothiocyanate (FITC) avidin and myeloperoxidase (MPO) staining. Moreover, the brain stem was harvested, and central nervous activation was investigated by Fos immunochemistry in both the nucleus of the solitary tract (NTS) and the area postrema (AP). Data are presented as mean ± SEM, and a p < 0.05 was considered statistically significant. KEY RESULTS Three hour experiments revealed no significant differences between all experimental groups, except MPO staining: 3 h after abdominal surgery, there were significantly more MPO-positive cells in vagotomized S-POI animals compared to sham-vagotomized S-POI animals (26.7 ± 7.1 vs. 5.1 ± 2.4, p < 0.01). Nine hour postoperatively intramuscular mast cells (IMMC) were significantly decreased in the intestinal muscularis of V/POI animals compared to SV/POI animals (1.5 ± 0.3 vs. 5.9 ± 0.2, p < 0.05), while MPO-positive cells were increased in V/POI animals compared to SV/POI animals (713.2 ± 99.4 vs. 46.9 ± 5.8, p < 0.05). There were less Fos-positive cells in the NTS of V/POI animals compared to SV/POI animals (64.7 ± 7.8 vs. 132.8 ± 23.9, p < 0.05) and more Fos-positive cells in the AP of V/POI animals compared to SV/POI animals 9 h postoperatively (38.0 ± 2.0 vs. 13.7 ± 0.9, p < 0.001). CONCLUSIONS AND INTERFERENCES Afferent nerve signaling to the central nervous system during the development of early POI seems to be mediated mainly via the vagus nerve and to a lesser degree via systemic circulation. During the early hours of POI, the intestinal immune response may be attenuated by vagal modulation, suggesting interactions between the central nervous system and the intestine.
Collapse
Affiliation(s)
- Martina Brandlhuber
- Department of Radiology, Grosshadern Clinic, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Petra Benhaqi
- Center for Human Genetics and Laboratory Diagnostics, Medical Labs Martinsried, Martinsried, Germany
| | | | - Vanessa Koliogiannis
- Department of Radiology, Grosshadern Clinic, Ludwig-Maximilians-University Munich, Munich, Germany
| | | | - Mario H Mueller
- Department of Surgery and Minimal-Invasive Surgery, Vivantes Klinikum Neukölln, Berlin, Germany
| | - Martin E Kreis
- Department of General and Visceral Surgery, Charité University Medicine Berlin, Campus Benjamin Franklin, Berlin, Germany
| |
Collapse
|
54
|
Xiao J, Niu J, Xu B, Zhang R, Zhang M, Zhang N, Xu K, Zhang Q, Chen D, Shi Y, Fang Q, Li N. NOP01, a NOP receptor agonist, produced potent and peripherally restricted antinociception in a formalin-induced mouse orofacial pain model. Neuropeptides 2022; 91:102212. [PMID: 34826712 DOI: 10.1016/j.npep.2021.102212] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/25/2021] [Accepted: 11/16/2021] [Indexed: 11/20/2022]
Abstract
Orofacial pain is one of the most common medical challenges. A preliminary report indicates that the NOP receptor may act as a therapeutic target in orofacial pain. Previous studies have shown that [(pF)Phe4, Aib7, Aib11, Arg14, Lys15]N/OFQ-NH2 (NOP01) functions as a potent NOP receptor peptide agonist. This work aims to investigate the antinociception of NOP01 and its possible action mechanisms in a formalin-induced mouse orofacial pain model at different levels. Our results demonstrated that local, intraperitoneal (i.p.) or intrathecal (i.t.) injection of NOP01 produced dose-related antinociception in both phases of the formalin pain, which could be inhibited by the NOP receptor antagonist but not the classical opioid receptor antagonist. Furthermore, the antinociception induced by systemic NOP01 was blocked by local but not spinal pretreatment with the NOP receptor antagonist, suggesting the involvement of the peripheral NOP receptor in NOP01-induced systemic antinociception. Moreover, local injection of NOP01 markedly suppressed the expression of c-Fos protein induced by formalin in ipsilateral trigeminal ganglion (TG) neurons. In conclusion, this work suggests that NOP01 exerts significant antinociception on orofacial pain at both peripheral and spinal levels via the NOP receptor. Notably, NOP01 cannot readily penetrate the blood-brain barrier. Thus, NOP01 may behave as a potential compound for developing peripherally restricted analgesics.
Collapse
Affiliation(s)
- Jian Xiao
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, PR China
| | - Jiandong Niu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, PR China
| | - Biao Xu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, PR China
| | - Run Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, PR China
| | - Mengna Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, PR China
| | - Nan Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, PR China
| | - Kangtai Xu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, PR China
| | - Qinqin Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, PR China
| | - Dan Chen
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, PR China
| | - Yonghang Shi
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, PR China
| | - Quan Fang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, PR China.
| | - Ning Li
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, PR China.
| |
Collapse
|
55
|
Inflammatory Stress Induced by Intraperitoneal Injection of LPS Increases Phoenixin Expression and Activity in Distinct Rat Brain Nuclei. Brain Sci 2022; 12:brainsci12020135. [PMID: 35203899 PMCID: PMC8870310 DOI: 10.3390/brainsci12020135] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/12/2022] [Accepted: 01/17/2022] [Indexed: 11/17/2022] Open
Abstract
Due to phoenixin’s role in restraint stress and glucocorticoid stress, as well as its recently shown effects on the inflammasome, we aimed to investigate the effects of lipopolysaccharide (LPS)-induced inflammatory stress on the activity of brain nuclei-expressing phoenixin. Male Sprague Dawley rats (n = 6/group) were intraperitoneally injected with either LPS or control (saline). Brains were processed for c-Fos and phoenixin immunohistochemistry and the resulting slides were evaluated using ImageJ software. c-Fos was counted and phoenixin was evaluated using densitometry. LPS stress significantly increased c-Fos expression in the central amygdaloid nucleus (CeM, 7.2-fold), supraoptic nucleus (SON, 34.8 ± 17.3 vs. 0.0 ± 0.0), arcuate nucleus (Arc, 4.9-fold), raphe pallidus (RPa, 5.1-fold), bed nucleus of the stria terminalis (BSt, 5.9-fold), dorsal motor nucleus of the vagus nerve (DMN, 89-fold), and medial part of the nucleus of the solitary tract (mNTS, 121-fold) compared to the control-injected group (p < 0.05). Phoenixin expression also significantly increased in the CeM (1.2-fold), SON (1.5-fold), RPa (1.3-fold), DMN (1.3-fold), and mNTS (1.9-fold, p < 0.05), leading to a positive correlation between c-Fos and phoenixin in the RPa, BSt, and mNTS (p < 0.05). In conclusion, LPS stress induces a significant increase in activity in phoenixin immunoreactive brain nuclei that is distinctively different from restraint stress.
Collapse
|
56
|
Keringer P, Furedi N, Gaszner B, Miko A, Pakai E, Fekete K, Olah E, Kelava L, Romanovsky AA, Rumbus Z, Garami A. The hyperthermic effect of central cholecystokinin is mediated by the cyclooxygenase-2 pathway. Am J Physiol Endocrinol Metab 2022; 322:E10-E23. [PMID: 34779255 DOI: 10.1152/ajpendo.00223.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cholecystokinin (CCK) increases core body temperature via CCK2 receptors when administered intracerebroventricularly (icv). The mechanisms of CCK-induced hyperthermia are unknown, and it is also unknown whether CCK contributes to the fever response to systemic inflammation. We studied the interaction between central CCK signaling and the cyclooxygenase (COX) pathway. Body temperature was measured in adult male Wistar rats pretreated with intraperitoneal infusion of the nonselective COX enzyme inhibitor metamizol (120 mg/kg) or a selective COX-2 inhibitor, meloxicam, or etoricoxib (10 mg/kg for both) and, 30 min later, treated with intracerebroventricular CCK (1.7 µg/kg). In separate experiments, CCK-induced neuronal activation (with and without COX inhibition) was studied in thermoregulation- and feeding-related nuclei with c-Fos immunohistochemistry. CCK increased body temperature by ∼0.4°C from 10 min postinfusion, which was attenuated by metamizol. CCK reduced the number of c-Fos-positive cells in the median preoptic area (by ∼70%) but increased it in the dorsal hypothalamic area and in the rostral raphe pallidus (by ∼50% in both); all these changes were completely blocked with metamizol. In contrast, CCK-induced satiety and neuronal activation in the ventromedial hypothalamus were not influenced by metamizol. CCK-induced hyperthermia was also completely blocked with both selective COX-2 inhibitors studied. Finally, the CCK2 receptor antagonist YM022 (10 µg/kg icv) attenuated the late phases of fever induced by bacterial lipopolysaccharide (10 µg/kg; intravenously). We conclude that centrally administered CCK causes hyperthermia through changes in the activity of "classical" thermoeffector pathways and that the activation of COX-2 is required for the development of this response.NEW & NOTEWORTHY An association between central cholecystokinin signaling and the cyclooxygenase-prostaglandin E pathway has been proposed but remained poorly understood. We show that the hyperthermic response to the central administration of cholecystokinin alters the neuronal activity within efferent thermoeffector pathways and that these effects are fully blocked by the inhibition of cyclooxygenase. We also show that the activation of cyclooxygenase-2 is required for the hyperthermic effect of cholecystokinin and that cholecystokinin is a modulator of endotoxin-induced fever.
Collapse
Affiliation(s)
- Patrik Keringer
- Department of Thermophysiology, Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Nora Furedi
- Department of Anatomy, Research Group for Mood Disorders, Centre for Neuroscience, Medical School and Szentagothai Research Centre, University of Pécs, Pécs, Hungary
| | - Balazs Gaszner
- Department of Anatomy, Research Group for Mood Disorders, Centre for Neuroscience, Medical School and Szentagothai Research Centre, University of Pécs, Pécs, Hungary
| | - Alexandra Miko
- Institute for Translational Medicine, Medical School and Szentagothai Research Centre, University of Pécs, Pécs, Hungary
| | - Eszter Pakai
- Department of Thermophysiology, Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Kata Fekete
- Department of Thermophysiology, Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Emoke Olah
- Department of Thermophysiology, Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Leonardo Kelava
- Department of Thermophysiology, Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | | | - Zoltan Rumbus
- Department of Thermophysiology, Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Andras Garami
- Department of Thermophysiology, Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| |
Collapse
|
57
|
Tesic V, Ciric J, Jovanovic Macura I, Zogovic N, Milanovic D, Kanazir S, Perovic M. Corticosterone and Glucocorticoid Receptor in the Cortex of Rats during Aging-The Effects of Long-Term Food Restriction. Nutrients 2021; 13:nu13124526. [PMID: 34960078 PMCID: PMC8703853 DOI: 10.3390/nu13124526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/09/2021] [Accepted: 12/10/2021] [Indexed: 11/16/2022] Open
Abstract
Numerous beneficial effects of food restriction on aging and age-related pathologies are well documented. It is also well-established that both short- and long-term food restriction regimens induce elevated circulating levels of glucocorticoids, stress-induced hormones produced by adrenal glands that can also exert deleterious effects on the brain. In the present study, we examined the effect of long-term food restriction on the glucocorticoid hormone/glucocorticoid receptor (GR) system in the cortex during aging, in 18- and 24-month-old rats. Corticosterone level was increased in the cortex of aged ad libitum-fed rats. Food restriction induced its further increase, accompanied with an increase in the level of 11β-hydroxysteroid dehydrogenase type 1. However, alterations in the level of GR phosphorylated at Ser232 were not detected in animals on food restriction, in line with unaltered CDK5 level, the decrease of Hsp90, and an increase in a negative regulator of GR function, FKBP51. Moreover, our data revealed that reduced food intake prevented age-related increase in the levels of NFκB, gfap, and bax, confirming its anti-inflammatory and anti-apoptotic effects. Along with an increase in the levels of c-fos, our study provides additional evidences that food restriction affects cortical responsiveness to glucocorticoids during aging.
Collapse
Affiliation(s)
- Vesna Tesic
- Department of Neurobiology, Institute for Biological Research “Sinisa Stankovic”—National Institute of Republic of Serbia, University of Belgrade, Bul. despota Stefana 142, 11060 Belgrade, Serbia; (V.T.); (J.C.); (I.J.M.); (D.M.); (M.P.)
| | - Jelena Ciric
- Department of Neurobiology, Institute for Biological Research “Sinisa Stankovic”—National Institute of Republic of Serbia, University of Belgrade, Bul. despota Stefana 142, 11060 Belgrade, Serbia; (V.T.); (J.C.); (I.J.M.); (D.M.); (M.P.)
| | - Irena Jovanovic Macura
- Department of Neurobiology, Institute for Biological Research “Sinisa Stankovic”—National Institute of Republic of Serbia, University of Belgrade, Bul. despota Stefana 142, 11060 Belgrade, Serbia; (V.T.); (J.C.); (I.J.M.); (D.M.); (M.P.)
| | - Nevena Zogovic
- Department of Neurophysiology, Institute for Biological Research “Sinisa Stankovic”—National Institute of Republic of Serbia, University of Belgrade, Bul. despota Stefana 142, 11060 Belgrade, Serbia;
| | - Desanka Milanovic
- Department of Neurobiology, Institute for Biological Research “Sinisa Stankovic”—National Institute of Republic of Serbia, University of Belgrade, Bul. despota Stefana 142, 11060 Belgrade, Serbia; (V.T.); (J.C.); (I.J.M.); (D.M.); (M.P.)
| | - Selma Kanazir
- Department of Neurobiology, Institute for Biological Research “Sinisa Stankovic”—National Institute of Republic of Serbia, University of Belgrade, Bul. despota Stefana 142, 11060 Belgrade, Serbia; (V.T.); (J.C.); (I.J.M.); (D.M.); (M.P.)
- Correspondence:
| | - Milka Perovic
- Department of Neurobiology, Institute for Biological Research “Sinisa Stankovic”—National Institute of Republic of Serbia, University of Belgrade, Bul. despota Stefana 142, 11060 Belgrade, Serbia; (V.T.); (J.C.); (I.J.M.); (D.M.); (M.P.)
| |
Collapse
|
58
|
Zhou M, Li D, Xie K, Xu L, Kong B, Wang X, Tang Y, Liu Y, Huang H. The short-chain fatty acid propionate improved ventricular electrical remodeling in a rat model with myocardial infarction. Food Funct 2021; 12:12580-12593. [PMID: 34813637 DOI: 10.1039/d1fo02040d] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The short-chain fatty acid (SCFA) propionate (C3), a microorganism metabolite produced by gut microbial fermentation, has parasympathetic-activation effects. The cardiac autonomic rebalancing strategy is considered as an important therapeutic approach to myocardial infarction (MI)-produced ventricular arrhythmias (VAs). Thus, our research was designed to clarify the potential functions of the SCFA propionate in VAs and cardiac electrophysiology in MI rats. A hundred adult Sprague-Dawley rats were allocated to four groups: the sham group (200 mM sodium chloride), the sham + C3 group (200 mM propionate), the MI group (200 mM sodium chloride) and the MI + C3 group (200 mM propionate). In comparison with the sham group, propionate significantly increased the parasympathetic components heart rate variability (HRV) and acetylcholine levels, prolonged cardiac repolarization, induced STAT3 phosphorylation and up-regulated the c-fos expression in nodose ganglia and solitary nucleus. Propionate intake reduced the susceptibility to VAs. MI induced by coronary ligation caused a significant increase in the sympathetic components HRV, abnormal repolarization, global repolarization dispersion, norepinephrine and inflammatory cytokines, reduction and redistribution of Connexin 43 in the infarcted border zone, and activation of NFκB, which were attenuated in the MI + C3 group. Oral propionate supplementation, as a nutritional intervention, protected the heart against MI-induced VAs and cardiac electrophysiology instability partly by parasympathetic activation based on the gut-brain axis.
Collapse
Affiliation(s)
- Mingmin Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China. .,Cardiovascular Research Institute of Wuhan University, Wuhan, China. .,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Diwen Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China. .,Cardiovascular Research Institute of Wuhan University, Wuhan, China. .,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Ke Xie
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China. .,Cardiovascular Research Institute of Wuhan University, Wuhan, China. .,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Liao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China. .,Cardiovascular Research Institute of Wuhan University, Wuhan, China. .,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Bin Kong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China. .,Cardiovascular Research Institute of Wuhan University, Wuhan, China. .,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Xi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China. .,Cardiovascular Research Institute of Wuhan University, Wuhan, China. .,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yanhong Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China. .,Cardiovascular Research Institute of Wuhan University, Wuhan, China. .,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yu Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China. .,Cardiovascular Research Institute of Wuhan University, Wuhan, China. .,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - He Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China. .,Cardiovascular Research Institute of Wuhan University, Wuhan, China. .,Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
59
|
Balcerek E, Włodkowska U, Czajkowski R. Retrosplenial cortex in spatial memory: focus on immediate early genes mapping. Mol Brain 2021; 14:172. [PMID: 34863215 PMCID: PMC8642902 DOI: 10.1186/s13041-021-00880-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 11/10/2021] [Indexed: 11/10/2022] Open
Abstract
The ability to form, retrieve and update autobiographical memories is one of the most fascinating features of human behavior. Spatial memory, the ability to remember the layout of the external environment and to navigate within its boundaries, is closely related to the autobiographical memory domain. It is served by an overlapping brain circuit, centered around the hippocampus (HPC) where the cognitive map index is stored. Apart from the hippocampus, several cortical structures participate in this process. Their relative contribution is a subject of intense research in both humans and animal models. One of the most widely studied regions is the retrosplenial cortex (RSC), an area in the parietal lobe densely interconnected with the hippocampal formation. Several methodological approaches have been established over decades in order to investigate the cortical aspects of memory. One of the most successful techniques is based on the analysis of brain expression patterns of the immediate early genes (IEGs). The common feature of this diverse group of genes is fast upregulation of their mRNA translation upon physiologically relevant stimulus. In the central nervous system they are rapidly triggered by neuronal activity and plasticity during learning. There is a widely accepted consensus that their expression level corresponds to the engagement of individual neurons in the formation of memory trace. Imaging of the IEGs might therefore provide a picture of an emerging memory engram. In this review we present the overview of IEG mapping studies of retrosplenial cortex in rodent models. We begin with classical techniques, immunohistochemical detection of protein and fluorescent in situ hybridization of mRNA. We then proceed to advanced methods where fluorescent genetically encoded IEG reporters are chronically followed in vivo during memory formation. We end with a combination of genetic IEG labelling and optogenetic approach, where the activity of the entire engram is manipulated. We finally present a hypothesis that attempts to unify our current state of knowledge about the function of RSC.
Collapse
Affiliation(s)
- Edyta Balcerek
- Laboratory of Spatial Memory, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3, 02-093, Warsaw, Poland
| | - Urszula Włodkowska
- Laboratory of Spatial Memory, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3, 02-093, Warsaw, Poland
| | - Rafał Czajkowski
- Laboratory of Spatial Memory, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3, 02-093, Warsaw, Poland.
| |
Collapse
|
60
|
Ambler M, Hitrec T, Pickering A. Turn it off and on again: characteristics and control of torpor. Wellcome Open Res 2021; 6:313. [DOI: 10.12688/wellcomeopenres.17379.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/12/2021] [Indexed: 11/20/2022] Open
Abstract
Torpor is a hypothermic, hypoactive, hypometabolic state entered into by a wide range of animals in response to environmental challenge. This review summarises the current understanding of torpor. We start by describing the characteristics of the wide-ranging physiological adaptations associated with torpor. Next follows a discussion of thermoregulation, control of food intake and energy expenditure, and the interactions of sleep and thermoregulation, with particular emphasis on how those processes pertain to torpor. We move on to take a critical view of the evidence for the systems that control torpor entry, including both the efferent circulating factors that signal the need for torpor, and the central processes that orchestrate it. Finally, we consider how the putative circuits responsible for torpor induction integrate with the established understanding of thermoregulation under non-torpid conditions and highlight important areas of uncertainty for future studies.
Collapse
|
61
|
Okuda T, Osako Y, Hidaka C, Nishihara M, Young LJ, Mitsui S, Yuri K. Separation from a bonded partner alters neural response to inflammatory pain in monogamous rodents. Behav Brain Res 2021; 418:113650. [PMID: 34748865 DOI: 10.1016/j.bbr.2021.113650] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 10/04/2021] [Accepted: 10/28/2021] [Indexed: 11/02/2022]
Abstract
Pain experience is known to be modified by social factors, but the brain mechanisms remain unspecified. We recently established an animal model of social stress-induced hyperalgesia (SSIH) using a socially monogamous rodent, the prairie vole, in which males separated from their female partners (loss males) became anxious and displayed exacerbated inflammatory pain behaviors compared to males with partners (paired males). In the present study, to explore the neural pathways involved in SSIH, a difference in neuronal activation in pain-related brain regions, or "pain matrix", during inflammatory pain between paired and loss males was detected using Fos immunoreactivity (Fos-ir). Males were paired with a female and pair bonding was confirmed in all subjects using a partner preference test. During formalin-induced inflammatory pain, both paired and loss males showed a significant induction of Fos-ir throughout the analyzed pain matrix components compared to basal condition (without injection), and no group differences in immunoreactivity were found among the injected males in many brain regions. However, the loss males had significantly lower Fos-ir following inflammatory pain in the medial prefrontal cortex and nucleus accumbens shell than the paired males, even though base Fos-ir levels were comparable between groups. Notably, both regions with different Fos-ir are major components of the dopamine and oxytocin systems, which play critical roles in both pair bonding and pain regulation. The present results suggest the possibility that pain exacerbation by social stress emerges through alteration of signaling in social brain circuitry.
Collapse
Affiliation(s)
- Takahiro Okuda
- Department of Neurobiology and Anatomy, Kochi Medical School, Kochi University, Oko-cho, Nankoku, Kochi 783-8505, Japan; Department of Physical Therapy, Tosa Rehabilitation College, Otsu, Ohtsu, Kochi 781-5103, Japan.
| | - Yoji Osako
- Department of Neurobiology and Anatomy, Kochi Medical School, Kochi University, Oko-cho, Nankoku, Kochi 783-8505, Japan
| | - Chiharu Hidaka
- Department of Neurobiology and Anatomy, Kochi Medical School, Kochi University, Oko-cho, Nankoku, Kochi 783-8505, Japan
| | - Makoto Nishihara
- Multidisciplinary Pain Centre, Aichi Medical University, School of Medicine, 21 Karimata, Nagakute, Aichi, 480-1195, Japan
| | - Larry J Young
- Silvio O. Conte Center for Oxytocin and Social Cognition, Center for Translational Social Neuroscience, Department of Psychiatry and Behavioral Sciences, Yerkes National Primate Center, Emory University School of Medicine, 954 Gatewood Rd. Atlanta, GA 30322, USA; Center for Social Neural Networks, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8577, Japan
| | - Shinichi Mitsui
- Department of Rehabilitation Sciences, Gunma University Graduate School of Health Sciences, 3-39-22 Showa-machi, Maebashi, Gunma 371-8514, Japan
| | - Kazunari Yuri
- Department of Neurobiology and Anatomy, Kochi Medical School, Kochi University, Oko-cho, Nankoku, Kochi 783-8505, Japan
| |
Collapse
|
62
|
Bryan de la Peña J, Kunder N, Lou TF, Chase R, Stanowick A, Barragan-Iglesias P, Pancrazio JJ, Campbell ZT. A Role for Translational Regulation by S6 Kinase and a Downstream Target in Inflammatory Pain. Br J Pharmacol 2021; 178:4675-4690. [PMID: 34355805 DOI: 10.1111/bph.15646] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Translational controls pervade neurobiology. Nociceptors play an integral role in the detection and propagation of pain signals. Nociceptors can undergo persistent changes in their intrinsic excitability. Pharmacologic disruption of nascent protein synthesis diminishes acute and chronic forms of pain-associated behaviors. Yet, the targets of translational controls that facilitate plasticity in nociceptors are unclear. EXPERIMENTAL APPROACH We used ribosome profiling to probe the translational landscape in DRG neurons after treatment of the inflammatory mediators NGF and IL-6. We validated the expression dynamics of c-Fos using immunoblotting and immunohistochemistry. Given that inflammation is known to stimulate mTOR signaling, we reasoned that downstream factors (e.g., ribosomal protein S6 kinase 1, S6K1) might control c-Fos levels. We utilized small-molecule inhibitors of S6K1 (DG2) or c-Fos (T-5224) to probe their effects on nociceptor activity in vitro using multi-electrode arrays (MEAs) and pain behavior in vivo using a hyperalgesic priming model. KEY RESULTS We demonstrate that c-Fos is expressed in sensory neurons. Inflammatory mediators that promote pain in both humans and rodents promote c-Fos translation. We demonstrate that the mTOR effector S6K1 is essential for c-Fos biosynthesis. Inhibition of S6K1 or c-Fos with small molecules diminish mechanical and thermal hypersensitivity in response to inflammatory cues. Additionally, both inhibitors reduce evoked nociceptor activity. CONCLUSION Our data reveal a novel role of S6K1 in modulating rapid response to inflammatory mediators, with c-Fos being one key downstream target. Targeting the S6 kinase pathway or c-Fos is an exciting new avenue for pain-modulating compounds.
Collapse
Affiliation(s)
- June Bryan de la Peña
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Nikesh Kunder
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Tzu-Fang Lou
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Rebecca Chase
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Alexander Stanowick
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Paulino Barragan-Iglesias
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA.,Department of Physiology and Pharmacology, Center for Basic Sciences, Autonomous University of Aguascalientes, Aguascalientes, Mexico
| | - Joseph J Pancrazio
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA.,Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA
| | - Zachary T Campbell
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, USA.,Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA.,Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA
| |
Collapse
|
63
|
Garcia-Lopez R, Pombero A, Estirado A, Geijo-Barrientos E, Martinez S. Interneuron Heterotopia in the Lis1 Mutant Mouse Cortex Underlies a Structural and Functional Schizophrenia-Like Phenotype. Front Cell Dev Biol 2021; 9:693919. [PMID: 34327202 PMCID: PMC8313859 DOI: 10.3389/fcell.2021.693919] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 06/16/2021] [Indexed: 11/24/2022] Open
Abstract
LIS1 is one of the principal genes related to Type I lissencephaly, a severe human brain malformation characterized by an abnormal neuronal migration in the cortex during embryonic development. This is clinically associated with epilepsy and cerebral palsy in severe cases, as well as a predisposition to developing mental disorders, in cases with a mild phenotype. Although genetic variations in the LIS1 gene have been associated with the development of schizophrenia, little is known about the underlying neurobiological mechanisms. We have studied how the Lis1 gene might cause deficits associated with the pathophysiology of schizophrenia using the Lis1/sLis1 murine model, which involves the deletion of the first coding exon of the Lis1 gene. Homozygous mice are not viable, but heterozygous animals present abnormal neuronal morphology, cortical dysplasia, and enhanced cortical excitability. We have observed reduced number of cells expressing GABA-synthesizing enzyme glutamic acid decarboxylase 67 (GAD67) in the hippocampus and the anterior cingulate area, as well as fewer parvalbumin-expressing cells in the anterior cingulate cortex in Lis1/sLis1 mutants compared to control mice. The cFOS protein expression (indicative of neuronal activity) in Lis1/sLis1 mice was higher in the medial prefrontal (mPFC), perirhinal (PERI), entorhinal (ENT), ectorhinal (ECT) cortices, and hippocampus compared to control mice. Our results suggest that deleting the first coding exon of the Lis1 gene might cause cortical anomalies associated with the pathophysiology of schizophrenia.
Collapse
Affiliation(s)
| | - Ana Pombero
- Instituto de Neurociencias, UMH-CSIC, Alicante, Spain
| | | | | | - Salvador Martinez
- Instituto de Neurociencias, UMH-CSIC, Alicante, Spain.,Centro de Investigación Biomédica En Red en Salud Mental-CIBERSAM-ISCIII, Valencia, Spain
| |
Collapse
|
64
|
Mapping excessive "disgust" in the brain: Ventral pallidum inactivation recruits distributed circuitry to make sweetness "disgusting". COGNITIVE AFFECTIVE & BEHAVIORAL NEUROSCIENCE 2021; 20:141-159. [PMID: 31836960 DOI: 10.3758/s13415-019-00758-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The ventral pallidum (VP) is an important structure in processing reward. The VP may be the only brain structure where localized lesions in rats replace normal facial "liking" expressions to sweetness with excessive "disgust" reactions, such as gapes and chin rubs, that are normally reserved for unpalatable tastes. The posterior half of the VP (pVP) contains a hedonic hot spot where opioid or related neurochemical stimulations can amplify positive "liking" reactions to sweet taste. This is the same site where lesions or pharmacological inactivations replace positive hedonic reactions to sucrose with intense negative "disgust." In the present study, we aimed to identify brain networks recruited by pVP inactivation to generate excessive "disgust," using neuronal Fos expression as a marker of neurobiological activation. Microinjections in pVP of inhibitory GABAA/B agonists (muscimol and baclofen) caused rats to exhibit excessive "disgust" reactions to sucrose. Excessive "disgust" was accompanied by recruitment of neural Fos activation in several subcortical structures, including the posterior medial shell of nucleus accumbens (which also contains another GABAergic "disgust"-inducing "hedonic cold spot"), the bed nucleus of stria terminalis, lateral habenula, hypothalamus, and midbrain ventral tegmentum. Fos suppression was found in cortical limbic regions, including previously identified hedonic hot spots in the anteromedial orbitofrontal cortex and posterior insula. Finally, in addition to inducing excessive "disgust," pVP inactivation abolished motivational "wanting" to eat palatable food, reduced positive social interactions, and reordered sensorimotor relations. Our findings identify potential "disgust" generators in the brain that are released into excitation by pVP inhibition and may serve as targets for future research.
Collapse
|
65
|
Neural substrates involved in the cognitive information processing in teleost fish. Anim Cogn 2021; 24:923-946. [PMID: 33907938 PMCID: PMC8360893 DOI: 10.1007/s10071-021-01514-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 02/25/2021] [Accepted: 03/06/2021] [Indexed: 02/04/2023]
Abstract
Over the last few decades, it has been shown that fish, comprising the largest group of vertebrates and in many respects one of the least well studied, possess many cognitive abilities comparable to those of birds and mammals. Despite a plethora of behavioural studies assessing cognition abilities and an abundance of neuroanatomical studies, only few studies have aimed to or in fact identified the neural substrates involved in the processing of cognitive information. In this review, an overview of the currently available studies addressing the joint research topics of cognitive behaviour and neuroscience in teleosts (and elasmobranchs wherever possible) is provided, primarily focusing on two fundamentally different but complementary approaches, i.e. ablation studies and Immediate Early Gene (IEG) analyses. More recently, the latter technique has become one of the most promising methods to visualize neuronal populations activated in specific brain areas, both during a variety of cognitive as well as non-cognition-related tasks. While IEG studies may be more elegant and potentially easier to conduct, only lesion studies can help researchers find out what information animals can learn or recall prior to and following ablation of a particular brain area.
Collapse
|
66
|
Chen H, Lv P, Liu Z, Chen W, Yao Y, Liu C, Cao Q, Zhou H. Preliminary study on the function of TMEM50A and its correlation with the RH genes. Transfus Med 2021; 31:277-285. [PMID: 33899290 DOI: 10.1111/tme.12778] [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: 06/04/2020] [Revised: 03/03/2021] [Accepted: 04/11/2021] [Indexed: 11/29/2022]
Abstract
OBJECTIVES The purpose of this study was to investigate the association and impact of TMEM50A on RH genes activity and function. BACKGROUND SMP1 is located on chromosome 1p36.11 in the RH gene locus, between the RHD and RHCE gene, where its position may be linked to RH haplotypes and contribute to selective pressures regarding certain RH haplotypes. TMEM50A is encoded by the SMP1 located in the intergenic region of RH, its influence on the function of the RH genes remains unclear. METHODS The expression of TMEM50A was regulated by transfection of plasmid and siRNA in K562 cell model. Western blot and real-time PCR were used to detect possible expression changes in the RH. The ammonium transport function of cells was monitored using pH-sensitive dye, while transcriptome sequencing was used to predict the potential function of TMEM50A. RESULTS The overexpression of TMEM50A significantly up-regulated RHCE gene activity (63.56%). The inhibition of TMEM50A resulted in significantly decreased RHCE (41.82%) and RHD expression (27.35%). Compared to control group, there was no significant change in the NH4 + transport function of cells in the overexpressed TMEM50A group. Transcriptome analysis showed that TMEM50A not only affected the transcription of target gene through splicing activities, but also played a role in the development of embryonic nervous system. CONCLUSIONS TMEM50A may regulate the expression of RH gene by affecting the stability of RH mRNA through splicing function. It speculates that TMEM50A may play an important role in the development of embryonic nervous system.
Collapse
Affiliation(s)
- Hongtian Chen
- Department of Blood Transfusion, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Piao Lv
- Department of Blood Transfusion, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ziwei Liu
- Department of Blood Transfusion, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wanjun Chen
- Department of Blood Transfusion, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuan Yao
- Department of Blood Transfusion, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chixiang Liu
- Department of Blood Transfusion, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qiong Cao
- Department of Blood Transfusion, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Huayou Zhou
- Department of Blood Transfusion, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
67
|
Takeda K, Ono H, Ishikawa K, Ohno T, Kumagai J, Ochiai H, Matumoto A, Yokoh H, Maezawa Y, Yokote K. Central administration of sodium-glucose cotransporter-2 inhibitors increases food intake involving adenosine monophosphate-activated protein kinase phosphorylation in the lateral hypothalamus in healthy rats. BMJ Open Diabetes Res Care 2021; 9:9/1/e002104. [PMID: 33879516 PMCID: PMC8061802 DOI: 10.1136/bmjdrc-2020-002104] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/03/2021] [Accepted: 03/21/2021] [Indexed: 01/14/2023] Open
Abstract
INTRODUCTION Sodium glucose cotransporter-2 (SGLT2) inhibitors are widely used for diabetes treatment. Although SGLT2 inhibitors have been clinically observed to increase food intake, roles or even the presence of SGLT2 in the central nervous system (CNS) has not been established. We aimed to elucidate potential functions of SGLT2 in the CNS, and the effects of CNS-targeted SGLT2 inhibitors on food intake. RESEARCH DESIGN AND METHODS We administered three kinds of SGLT2 inhibitors, tofogliflozin, dapagliflozin, and empagliflozin, into the lateral ventricle (LV) in rats and evaluated their effects on food intake. We also evaluated the effects of tofogliflozin administration in the third (3V) and fourth ventricle (4V). Intraperitoneal administration of liraglutide, a glucagon-like peptide-1 (GLP-1) receptor agonist known to suppress food intake, was combined with central tofogliflozin to elucidate whether GLP-1 signaling antagonizes the effect of central SGLT2 inhibitors on food intake. To elucidate potential molecular mechanisms mediating changes in feeding, hypothalamic areas associated with food intake regulation were harvested and analyzed after intracerebroventricular administration (ICV) of tofogliflozin. RESULTS Bolus ICV injection of tofogliflozin induced a robust increase in food intake starting at 1.5 hours postinjection, and lasting for 5 days. No effect was observed when the same dose of tofogliflozin was administered intraperitoneally. ICV dapagliflozin and empagliflozin significantly enhanced food intake, although the strength of these effects varied among drugs. Food intake was most markedly enhanced when tofogliflozin was infused into the LV. Fewer or no effects were observed with infusion into the 3V or 4V, respectively. Systemic administration of liraglutide suppressed the effect of ICV tofogliflozin on food intake. ICV tofogliflozin increased phosphorylation of AMPK and c-fos expression in the lateral hypothalamus. CONCLUSIONS SGLT2 inhibitors in the CNS increase food intake. SGLT2 activity in the CNS may regulate food intake through AMPK phosphorylation in the lateral hypothalamic area.
Collapse
Affiliation(s)
- Kenji Takeda
- Department of Endocrinology, Hematology, and Gerontology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Hiraku Ono
- Department of Endocrinology, Hematology, and Gerontology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Ko Ishikawa
- Department of Endocrinology, Hematology, and Gerontology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Tomohiro Ohno
- Department of Endocrinology, Hematology, and Gerontology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Jin Kumagai
- Department of Endocrinology, Hematology, and Gerontology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Hidetoshi Ochiai
- Department of Endocrinology, Hematology, and Gerontology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Ai Matumoto
- Department of Endocrinology, Hematology, and Gerontology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Hidetaka Yokoh
- Department of Endocrinology, Hematology, and Gerontology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Yoshiro Maezawa
- Department of Endocrinology, Hematology, and Gerontology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Koutaro Yokote
- Department of Endocrinology, Hematology, and Gerontology, Chiba University Graduate School of Medicine, Chiba, Japan
| |
Collapse
|
68
|
Alachkar A, Lotfy M, Adeghate E, Łażewska D, Kieć-Kononowicz K, Sadek B. Ameliorating effects of histamine H3 receptor antagonist E177 on acute pentylenetetrazole-induced memory impairments in rats. Behav Brain Res 2021; 405:113193. [PMID: 33626390 DOI: 10.1016/j.bbr.2021.113193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 02/14/2021] [Accepted: 02/14/2021] [Indexed: 12/14/2022]
Abstract
Histamine H3 receptors (H3Rs) are involved in several neuropsychiatric diseases including epilepsy. Therefore, the effects of H3R antagonist E177 (5 and 10 mg/kg, intraperitoneal (i.p.)) were evaluated on acute pentylenetetrazole (PTZ)-induced memory impairments, oxidative stress levels (glutathione (GSH), malondialdehyde (MDA), catalase (CAT), and superoxide dismutase (SOD)), various brain neurotransmitters (histamine (HA), acetylcholine (ACh), γ-aminobutyric acid (GABA)), and glutamate (Glu), acetylcholine esterase (AChE) activity, and c-fos protein expression in rats. E177 (5 and 10 mg/kg, i.p.) significantly prolonged step-through latency (STL) time in single-trial passive avoidance paradigm (STPAP), and shortened transfer latency time (TLT) in elevated plus maze paradigm (EPMP) (all P < 0.05). Moreover, and in the hippocampus of PTZ-treated animals, E177 mitigated abnormal levels of AChE activity, ACh and HA (all P < 0.05), but failed to modify brain levels of GABA and Glu. Furthermore, E177 alleviated hippocampal oxidative stress by significantly decreasing the elevated levels of MDA, and increasing the abnormally decreased level of GSH (all P < 0.05). Furthermore, E177 reduced elevated levels of hippocampal c-fos protein expression in hippocampal tissues of PTZ-treated animals (all P < 0.05). The observed results propose the potential of H3R antagonist E177 with an added advantage of avoiding cognitive impairment, emphasizing the H3Rs as a prospective target for future pharmacological management of epilepsy with associated memory impairments.
Collapse
Affiliation(s)
- Alaa Alachkar
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, 17666, United Arab Emirates; Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, P.O. Box 17666, Abu Dhabi, United Arab Emirates
| | - Mohamed Lotfy
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, 17666, United Arab Emirates
| | - Ernest Adeghate
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, P.O. Box 17666, Abu Dhabi, United Arab Emirates; Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, 17666, United Arab Emirates
| | - Dorota Łażewska
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Technology and Biotechnology of Drugs, Medyczna 9 St., 30-688, Kraków, Poland
| | - Katarzyna Kieć-Kononowicz
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Technology and Biotechnology of Drugs, Medyczna 9 St., 30-688, Kraków, Poland
| | - Bassem Sadek
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, 17666, United Arab Emirates; Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, P.O. Box 17666, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
69
|
Trojanowski NF, Bottorff J, Turrigiano GG. Activity labeling in vivo using CaMPARI2 reveals intrinsic and synaptic differences between neurons with high and low firing rate set points. Neuron 2021; 109:663-676.e5. [PMID: 33333001 PMCID: PMC7897300 DOI: 10.1016/j.neuron.2020.11.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 10/27/2020] [Accepted: 11/24/2020] [Indexed: 11/25/2022]
Abstract
Neocortical pyramidal neurons regulate firing around a stable mean firing rate (FR) that can differ by orders of magnitude between neurons, but the factors that determine where individual neurons sit within this broad FR distribution are not understood. To access low- and high-FR neurons for ex vivo analysis, we used Ca2+- and UV-dependent photoconversion of CaMPARI2 in vivo to permanently label neurons according to mean FR. CaMPARI2 photoconversion was correlated with immediate early gene expression and higher FRs ex vivo and tracked the drop and rebound in ensemble mean FR induced by prolonged monocular deprivation. High-activity L4 pyramidal neurons had greater intrinsic excitability and recurrent excitatory synaptic strength, while E/I ratio, local output strength, and local connection probability were not different. Thus, in L4 pyramidal neurons (considered a single transcriptional cell type), a broad mean FR distribution is achieved through graded differences in both intrinsic and synaptic properties.
Collapse
Affiliation(s)
| | - Juliet Bottorff
- Department of Biology, Brandeis University, Waltham, MA 02453, USA
| | | |
Collapse
|
70
|
Hoffiz YC, Castillo-Ruiz A, Hall MAL, Hite TA, Gray JM, Cisternas CD, Cortes LR, Jacobs AJ, Forger NG. Birth elicits a conserved neuroendocrine response with implications for perinatal osmoregulation and neuronal cell death. Sci Rep 2021; 11:2335. [PMID: 33504846 PMCID: PMC7840942 DOI: 10.1038/s41598-021-81511-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 01/05/2021] [Indexed: 12/17/2022] Open
Abstract
Long-standing clinical findings report a dramatic surge of vasopressin in umbilical cord blood of the human neonate, but the neural underpinnings and function(s) of this phenomenon remain obscure. We studied neural activation in perinatal mice and rats, and found that birth triggers activation of the suprachiasmatic, supraoptic, and paraventricular nuclei of the hypothalamus. This was seen whether mice were born vaginally or via Cesarean section (C-section), and when birth timing was experimentally manipulated. Neuronal phenotyping showed that the activated neurons were predominantly vasopressinergic, and vasopressin mRNA increased fivefold in the hypothalamus during the 2–3 days before birth. Copeptin, a surrogate marker of vasopressin, was elevated 30-to 50-fold in plasma of perinatal mice, with higher levels after a vaginal than a C-section birth. We also found an acute decrease in plasma osmolality after a vaginal, but not C-section birth, suggesting that the difference in vasopressin release between birth modes is functionally meaningful. When vasopressin was administered centrally to newborns, we found an ~ 50% reduction in neuronal cell death in specific brain areas. Collectively, our results identify a conserved neuroendocrine response to birth that is sensitive to birth mode, and influences peripheral physiology and neurodevelopment.
Collapse
Affiliation(s)
- Yarely C Hoffiz
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30302, USA
| | | | - Megan A L Hall
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30302, USA
| | - Taylor A Hite
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30302, USA
| | - Jennifer M Gray
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30302, USA
| | - Carla D Cisternas
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30302, USA.,Instituto de Investigación Médica M Y M Ferreyra, INIMEC-CONICET-UNC, Córdoba, Argentina
| | - Laura R Cortes
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30302, USA
| | - Andrew J Jacobs
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30302, USA
| | - Nancy G Forger
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30302, USA.
| |
Collapse
|
71
|
Hanada R. The role of the RANKL/RANK/OPG system in the central nervous systems (CNS). J Bone Miner Metab 2021; 39:64-70. [PMID: 32888064 DOI: 10.1007/s00774-020-01143-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 08/11/2020] [Indexed: 12/18/2022]
Abstract
The receptor-activator of NF-κB ligand (RANKL) and its specific receptor RANK have essential roles in regulating bone metabolism and the immune system. Besides, the RANKL/RANK system plays important roles in multiple physiological and pathophysiological processes such as mammary gland development during pregnancy, cancer development, and bone metastasis. While it has long been known that RANKL and RANK are expressed in the central nervous system (CNS), the physiological roles of RANKL/RANK system in the CNS and the underlying molecular mechanisms have been elucidated recently. Over the last decade, several reports showed that the central RANKL/RANK system plays important roles in regulating body temperature, brain ischemia, autoimmune encephalopathy, feeding behavior, and energy metabolism. In this review, it is provided an updated information regarding the roles of RANKL/RANK system in the CNS.
Collapse
Affiliation(s)
- Reiko Hanada
- Department of Neurophysiology, Faculty of Medicine, Oita University, Idaigaoka 1-1, Yufu City, Oita, 879-5593, Japan.
| |
Collapse
|
72
|
Pérez-Revuelta L, Téllez de Meneses PG, López M, Briñón JG, Weruaga E, Díaz D, Alonso JR. Secretagogin expression in the mouse olfactory bulb under sensory impairments. Sci Rep 2020; 10:21533. [PMID: 33299042 PMCID: PMC7726155 DOI: 10.1038/s41598-020-78499-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 11/24/2020] [Indexed: 12/04/2022] Open
Abstract
The interneurons of the olfactory bulb (OB) are characterized by the expression of different calcium-binding proteins, whose specific functions are not fully understood. This is the case of one of the most recently discovered, the secretagogin (SCGN), which is expressed in interneurons of the glomerular and the granule cell layers, but whose function in the olfactory pathway is still unknown. To address this question, we examined the distribution, generation and activity of SCGN-positive interneurons in the OB of two complementary models of olfactory impairments: Purkinje Cell Degeneration (PCD) and olfactory-deprived mice. Our results showed a significant increase in the density of SCGN-positive cells in the inframitral layers of olfactory-deprived mice as compared to control animals. Moreover, BrdU analyses revealed that these additional SCGN-positive cells are not newly formed. Finally, the neuronal activity, estimated by c-Fos expression, increased in preexisting SCGN-positive interneurons of both deprived and PCD mice -being higher in the later- in comparison with control animals. Altogether, our results suggest that the OB possesses different compensatory mechanisms depending on the type of alteration. Particularly, the SCGN expression is dependent of olfactory stimuli and its function may be related to a compensation against a reduction in sensory inputs.
Collapse
Affiliation(s)
- L Pérez-Revuelta
- Laboratory of Neuronal Plasticity and Neurorepair, Institute for Neuroscience of Castile and Leon (INCyL), University of Salamanca, C/ Pintor Fernando Gallego, 1, 37007, Salamanca, Spain.,Institute of Biomedical Research of Salamanca, IBSAL, 37007, Salamanca, Spain
| | - P G Téllez de Meneses
- Laboratory of Neuronal Plasticity and Neurorepair, Institute for Neuroscience of Castile and Leon (INCyL), University of Salamanca, C/ Pintor Fernando Gallego, 1, 37007, Salamanca, Spain.,Institute of Biomedical Research of Salamanca, IBSAL, 37007, Salamanca, Spain
| | - M López
- Laboratory of Neuronal Plasticity and Neurorepair, Institute for Neuroscience of Castile and Leon (INCyL), University of Salamanca, C/ Pintor Fernando Gallego, 1, 37007, Salamanca, Spain.,Institute of Biomedical Research of Salamanca, IBSAL, 37007, Salamanca, Spain
| | - J G Briñón
- Laboratory of Neuronal Plasticity and Neurorepair, Institute for Neuroscience of Castile and Leon (INCyL), University of Salamanca, C/ Pintor Fernando Gallego, 1, 37007, Salamanca, Spain.,Institute of Biomedical Research of Salamanca, IBSAL, 37007, Salamanca, Spain
| | - E Weruaga
- Laboratory of Neuronal Plasticity and Neurorepair, Institute for Neuroscience of Castile and Leon (INCyL), University of Salamanca, C/ Pintor Fernando Gallego, 1, 37007, Salamanca, Spain.,Institute of Biomedical Research of Salamanca, IBSAL, 37007, Salamanca, Spain
| | - D Díaz
- Laboratory of Neuronal Plasticity and Neurorepair, Institute for Neuroscience of Castile and Leon (INCyL), University of Salamanca, C/ Pintor Fernando Gallego, 1, 37007, Salamanca, Spain. .,Institute of Biomedical Research of Salamanca, IBSAL, 37007, Salamanca, Spain.
| | - J R Alonso
- Laboratory of Neuronal Plasticity and Neurorepair, Institute for Neuroscience of Castile and Leon (INCyL), University of Salamanca, C/ Pintor Fernando Gallego, 1, 37007, Salamanca, Spain.,Institute of Biomedical Research of Salamanca, IBSAL, 37007, Salamanca, Spain
| |
Collapse
|
73
|
Liu Y, Alkharabsheh A, Sun W. Hyperexcitability of the Nucleus Accumbens Is Involved in Noise-Induced Hyperacusis. Neural Plast 2020; 2020:8814858. [PMID: 33293947 PMCID: PMC7714561 DOI: 10.1155/2020/8814858] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 10/18/2020] [Accepted: 11/12/2020] [Indexed: 11/18/2022] Open
Abstract
Reduced tolerance to sound stimuli (hyperacusis) is commonly seen in tinnitus patients. Dysfunction of limbic systems, such as the nucleus accumbens (NAc), may be involved in emotional reactions to the sound stimuli in tinnitus patients. To study the functional changes in the NAc in hyperacusis, we have examined the neural activity changes of the NAc using c-Fos staining in an animal model of hyperacusis. The c-Fos staining was also examined in the medial geniculate nucleus (MGN), a central auditory pathway which has neural projections to the NAc. Postnatal rats (14 days) were exposed to loud noise (115 dB SPL, 4 hours for two consecutive days) to induce hyperacusis (n = 4). Rats without noise exposure were used as the controls (n = 4). After P35, rats in both groups were put in a behavioral training for sound detection. After they were trained to detect sound stimuli, their reaction time to noise bursts centered at 2 kHz (40-110 dB SPL) was measured. Rats in the noise group showed a significantly shorter reaction time than those in the control group to the noise bursts at high intensities, suggesting the noise exposure induced hyperacusis behavior. The c-Fos expressions in the NAc and the MGNs of the noise group were significantly higher than those of the control group. Our results suggested that early-age noise exposure caused hyperactivity in the NAc and the MGNs which may induce the loudness increase in these rats.
Collapse
Affiliation(s)
- Yuying Liu
- Department of Otorhinolaryngology-Head and Neck Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Shanghai, China 200080
| | - Ana''am Alkharabsheh
- Department of Hearing and Speech Sciences, University of Jordan, Queen Rania Al Abdallah St., Amman, Jordan 11942
| | - Wei Sun
- Department of Communicative Disorders and Sciences, Center for Hearing and Deafness, State University of New York at Buffalo, 137 Cary Hall, 3435 Main Street, Buffalo, NY 14214, USA
| |
Collapse
|
74
|
Guggenberger M, Engster KM, Hofmann T, Rose M, Stengel A, Kobelt P. Cholecystokinin and bombesin activate neuronatin neurons in the nucleus of the solitary tract. Brain Res 2020; 1746:147006. [DOI: 10.1016/j.brainres.2020.147006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 06/09/2020] [Accepted: 06/29/2020] [Indexed: 12/29/2022]
|
75
|
Franceschini A, Costantini I, Pavone FS, Silvestri L. Dissecting Neuronal Activation on a Brain-Wide Scale With Immediate Early Genes. Front Neurosci 2020; 14:569517. [PMID: 33192255 PMCID: PMC7645181 DOI: 10.3389/fnins.2020.569517] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 09/28/2020] [Indexed: 11/13/2022] Open
Abstract
Visualizing neuronal activation on a brain-wide scale yet with cellular resolution is a fundamental technical challenge for neuroscience. This would enable analyzing how different neuronal circuits are disrupted in pathology and how they could be rescued by pharmacological treatments. Although this goal would have appeared visionary a decade ago, recent technological advances make it eventually feasible. Here, we review the latest developments in the fields of genetics, sample preparation, imaging, and image analysis that could be combined to afford whole-brain cell-resolution activation mapping. We show how the different biochemical and optical methods have been coupled to study neuronal circuits at different spatial and temporal scales, and with cell-type specificity. The inventory of techniques presented here could be useful to find the tools best suited for a specific experiment. We envision that in the next years, mapping of neuronal activation could become routine in many laboratories, allowing dissecting the neuronal counterpart of behavior.
Collapse
Affiliation(s)
| | - Irene Costantini
- European Laboratory for Non-linear Spectroscopy (LENS), Sesto Fiorentino, Italy.,National Institute of Optics, National Research Council (INO-CNR), Sesto Fiorentino, Italy
| | - Francesco S Pavone
- European Laboratory for Non-linear Spectroscopy (LENS), Sesto Fiorentino, Italy.,National Institute of Optics, National Research Council (INO-CNR), Sesto Fiorentino, Italy.,Department of Physics and Astronomy, University of Florence, Florence, Italy
| | - Ludovico Silvestri
- European Laboratory for Non-linear Spectroscopy (LENS), Sesto Fiorentino, Italy.,National Institute of Optics, National Research Council (INO-CNR), Sesto Fiorentino, Italy.,Department of Physics and Astronomy, University of Florence, Florence, Italy
| |
Collapse
|
76
|
Immunohistochemical Evidence for Glutamatergic Regulation of Nesfatin-1 Neurons in the Rat Hypothalamus. Brain Sci 2020; 10:brainsci10090630. [PMID: 32932902 PMCID: PMC7564322 DOI: 10.3390/brainsci10090630] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 09/04/2020] [Accepted: 09/07/2020] [Indexed: 12/03/2022] Open
Abstract
Nesfatin-1, identified as an anorexigenic peptide, regulates the energy metabolism by suppressing food intake. The majority of nesfatin-1-synthesizing neurons are concentrated in various hypothalamic nuclei, especially in the supraoptic (SON), arcuate (ARC) and paraventricular nuclei (PVN). We tested the hypothesis that the glutamatergic system regulates nesfatin-1 neurons through glutamate receptors. Therefore, the first aim of the proposed studies was to examine effects of different glutamate agonists in the activation of nesfatin-1 neurons using c-Fos double immunohistochemical labeling. Experimental groups were formed containing male and female rats which received intraperitoneal injections of glutamate agonists kainic acid, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) and N-methyl-D-aspartate (NMDA) while the control rats received vehicle. The significant increase in the number of c-Fos-expressing nesfatin-1 neurons after agonist injections were observed both in female and male subjects and some of these effects were found to be sexually dimorphic. In addition, treatment with specific glutamate antagonists 6-cyano-7-nitroquinoxaline-2,3-dione (CNQX) or dizocilpine (MK-801) before each of the three agonist injections caused a statistically significant reduction in the number of activated nesfatin-1 neurons in the hypothalamic nuclei including supraoptic, paraventricular and arcuate nuclei. The second aim of the study was to determine the expression of glutamate receptor subunit proteins in the nesfatin-1 neurons by using a double immunofluorescence technique. The results showed that the glutamate receptor subunits, which may form homomeric or heteromeric functional receptor channels, were expressed in the nesfatin-1 neurons. In conclusion, the results of this study suggest that nesfatin-1 neurons respond to glutamatergic signals in the form of neuronal activation and that the glutamate receptors that are synthesized by nesfatin-1 neurons may participate in the glutamatergic regulation of these neurons.
Collapse
|
77
|
Rhinehart EM, Waldron M, Kelly-Quigley H, Zellers M, Turco A, Grisel JE. β-Endorphin and sex differentially modulate the response to EtOH in a site-specific manner. Brain Res 2020; 1741:146845. [DOI: 10.1016/j.brainres.2020.146845] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 04/15/2020] [Accepted: 04/16/2020] [Indexed: 02/06/2023]
|
78
|
Park J, Jin K, Sahasrabudhe A, Chiang PH, Maalouf JH, Koehler F, Rosenfeld D, Rao S, Tanaka T, Khudiyev T, Schiffer ZJ, Fink Y, Yizhar O, Manthiram K, Anikeeva P. In situ electrochemical generation of nitric oxide for neuronal modulation. NATURE NANOTECHNOLOGY 2020; 15:690-697. [PMID: 32601446 PMCID: PMC7415650 DOI: 10.1038/s41565-020-0701-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 04/27/2020] [Indexed: 05/07/2023]
Abstract
Understanding the function of nitric oxide, a lipophilic messenger in physiological processes across nervous, cardiovascular and immune systems, is currently impeded by the dearth of tools to deliver this gaseous molecule in situ to specific cells. To address this need, we have developed iron sulfide nanoclusters that catalyse nitric oxide generation from benign sodium nitrite in the presence of modest electric fields. Locally generated nitric oxide activates the nitric oxide-sensitive cation channel, transient receptor potential vanilloid family member 1 (TRPV1), and the latency of TRPV1-mediated Ca2+ responses can be controlled by varying the applied voltage. Integrating these electrocatalytic nanoclusters with multimaterial fibres allows nitric oxide-mediated neuronal interrogation in vivo. The in situ generation of nitric oxide in the ventral tegmental area with the electrocatalytic fibres evoked neuronal excitation in the targeted brain region and its excitatory projections. This nitric oxide generation platform may advance mechanistic studies of the role of nitric oxide in the nervous system and other organs.
Collapse
Affiliation(s)
- Jimin Park
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kyoungsuk Jin
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Atharva Sahasrabudhe
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Po-Han Chiang
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute of Biomedical Engineering, National Chiao Tung University, Taiwan, Taiwan
| | - Joseph H Maalouf
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Florian Koehler
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Dekel Rosenfeld
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Siyuan Rao
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Simons Center for Social Brain, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Tomo Tanaka
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- System Platform Research Laboratories, NEC Corporation, Tsukuba, Japan
| | - Tural Khudiyev
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Zachary J Schiffer
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yoel Fink
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ofer Yizhar
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Karthish Manthiram
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Polina Anikeeva
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA.
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
79
|
Bres EE, Safina D, Müller J, Bedner P, Yang H, Helluy X, Shchyglo O, Jansen S, Mark MD, Esser A, Steinhäuser C, Herlitze S, Pietrzik CU, Sirko S, Manahan-Vaughan D, Faissner A. Lipoprotein receptor loss in forebrain radial glia results in neurological deficits and severe seizures. Glia 2020; 68:2517-2549. [PMID: 32579270 DOI: 10.1002/glia.23869] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 05/19/2020] [Accepted: 05/22/2020] [Indexed: 02/06/2023]
Abstract
The Alzheimer disease-associated multifunctional low-density lipoprotein receptor-related protein-1 is expressed in the brain. Recent studies uncovered a role of this receptor for the appropriate functioning of neural stem cells, oligodendrocytes, and neurons. The constitutive knock-out (KO) of the receptor is embryonically lethal. To unravel the receptors' role in the developing brain we generated a mouse mutant by specifically targeting radial glia stem cells of the dorsal telencephalon. The low-density lipoprotein receptor-related protein-1 lineage-restricted KO female and male mice, in contrast to available models, developed a severe neurological phenotype with generalized seizures during early postnatal development. The mechanism leading to a buildup of hyperexcitability and emergence of seizures was traced to a failure in adequate astrocyte development and deteriorated postsynaptic density integrity. The detected impairments in the astrocytic lineage: precocious maturation, reactive gliosis, abolished tissue plasminogen activator uptake, and loss of functionality emphasize the importance of this glial cell type for synaptic signaling in the developing brain. Together, the obtained results highlight the relevance of astrocytic low-density lipoprotein receptor-related protein-1 for glutamatergic signaling in the context of neuron-glia interactions and stage this receptor as a contributing factor for epilepsy.
Collapse
Affiliation(s)
- Ewa E Bres
- Department of Cell Morphology and Molecular Neurobiology, Ruhr University Bochum, Bochum, Germany.,International Graduate School of Neuroscience, Ruhr University Bochum, Bochum, Germany
| | - Dina Safina
- Department of Cell Morphology and Molecular Neurobiology, Ruhr University Bochum, Bochum, Germany.,International Graduate School of Neuroscience, Ruhr University Bochum, Bochum, Germany
| | - Julia Müller
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Peter Bedner
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Honghong Yang
- Department of Neurophysiology, Medical Faculty, Ruhr University Bochum, Bochum, Germany
| | - Xavier Helluy
- Department of Neurophysiology, Medical Faculty, Ruhr University Bochum, Bochum, Germany.,Department of Psychology, Institute of Cognitive Neuroscience, Biopsychology, Ruhr University Bochum, Bochum, Germany
| | - Olena Shchyglo
- Department of Neurophysiology, Medical Faculty, Ruhr University Bochum, Bochum, Germany
| | - Stephan Jansen
- Department of Neurophysiology, Medical Faculty, Ruhr University Bochum, Bochum, Germany
| | - Melanie D Mark
- Behavioral Neuroscience, Ruhr University Bochum, Bochum, Germany
| | | | - Christian Steinhäuser
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Stefan Herlitze
- Department of General Zoology and Neurobiology, Ruhr University Bochum, Bochum, Germany
| | - Claus U Pietrzik
- Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Swetlana Sirko
- Department of Physiological Genomics, Biomedical Center (BMC), Ludwig-Maximilians University, Planegg-Martinsried, Germany.,Institute for Stem Cell Research, Helmholtz Zentrum Munich, Neuherberg, Germany
| | | | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
80
|
Poll S, Mittag M, Musacchio F, Justus LC, Giovannetti EA, Steffen J, Wagner J, Zohren L, Schoch S, Schmidt B, Jackson WS, Ehninger D, Fuhrmann M. Memory trace interference impairs recall in a mouse model of Alzheimer’s disease. Nat Neurosci 2020; 23:952-958. [DOI: 10.1038/s41593-020-0652-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 05/04/2020] [Indexed: 12/20/2022]
|
81
|
Friedrich T, Schalla MA, Lommel R, Goebel-Stengel M, Kobelt P, Rose M, Stengel A. Restraint stress increases the expression of phoenixin immunoreactivity in rat brain nuclei. Brain Res 2020; 1743:146904. [PMID: 32474019 DOI: 10.1016/j.brainres.2020.146904] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 05/01/2020] [Accepted: 05/22/2020] [Indexed: 12/14/2022]
Abstract
Phoenixin is a recently discovered peptide, which has been associated with reproduction, anxiety and food intake. Based on a considerable co-localization it has been linked to nesfatin-1, with a possible antagonistic mode of action. Since nesfatin-1 is known to play a role in anxiety and the response to stress, this study aims to investigate the effects of a well-established psychological stress model, restraint stress, on phoenixin-expressing brain nuclei and phoenixin expression in rats. Male Sprague-Dawley rats were subjected to restraint stress (n = 8) or left undisturbed (control, n = 6) and the brains processed for c-Fos- and phoenixin immunohistochemistry. The number of c-Fos expressing cells was counted and phoenixin expression assessed semiquantitatively. Restraint stress significantly increased c-Fos expression in the dorsal motor nucleus of vagus nerve (DMN, 52-fold, p < 0.001), raphe pallidus (RPa, 15-fold, p < 0.001), medial part of the nucleus of the solitary tract (mNTS, 16-fold, p < 0.001), central amygdaloid nucleus, medial division (CeM, 9-fold, p = 0.01), supraoptic nucleus (SON, 9-fold, p < 0.001) and the arcuate nucleus (Arc, 2.5-fold, p < 0.03) compared to control animals. Also phoenixin expression significantly increased in the DMN (17-fold, p < 0.001), RPa (2-fold, p < 0.001) and mNTS (1.6-fold, p < 0.001) with positive correlations between c-Fos and phoenixin (r = 0.74-0.85; p < 0.01) in these nuclei. This pattern of activation suggests an involvement of phoenixin in response to restraint stress. Whether phoenixin mediates stress effects or is activated in a counterbalancing fashion will have to be further investigated.
Collapse
Affiliation(s)
- T Friedrich
- Charité Center for Internal Medicine and Dermatology, Department for Psychosomatic Medicine, Charite - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - M A Schalla
- Charité Center for Internal Medicine and Dermatology, Department for Psychosomatic Medicine, Charite - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - R Lommel
- Charité Center for Internal Medicine and Dermatology, Department for Psychosomatic Medicine, Charite - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - M Goebel-Stengel
- Charité Center for Internal Medicine and Dermatology, Department for Psychosomatic Medicine, Charite - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany; Department of Internal Medicine, Helios Kliniken GmbH, Rottweil, Germany; Department of Psychosomatic Medicine and Psychotherapy, University Hospital Tübingen, Tübingen, Germany
| | - P Kobelt
- Charité Center for Internal Medicine and Dermatology, Department for Psychosomatic Medicine, Charite - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - M Rose
- Charité Center for Internal Medicine and Dermatology, Department for Psychosomatic Medicine, Charite - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - A Stengel
- Charité Center for Internal Medicine and Dermatology, Department for Psychosomatic Medicine, Charite - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany; Department of Psychosomatic Medicine and Psychotherapy, University Hospital Tübingen, Tübingen, Germany.
| |
Collapse
|
82
|
AUTS2 Regulation of Synapses for Proper Synaptic Inputs and Social Communication. iScience 2020; 23:101183. [PMID: 32498016 PMCID: PMC7267731 DOI: 10.1016/j.isci.2020.101183] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 04/30/2020] [Accepted: 05/15/2020] [Indexed: 01/11/2023] Open
Abstract
Impairments in synapse development are thought to cause numerous psychiatric disorders. Autism susceptibility candidate 2 (AUTS2) gene has been associated with various psychiatric disorders, such as autism and intellectual disabilities. Although roles for AUTS2 in neuronal migration and neuritogenesis have been reported, its involvement in synapse regulation remains unclear. In this study, we found that excitatory synapses were specifically increased in the Auts2-deficient primary cultured neurons as well as Auts2 mutant forebrains. Electrophysiological recordings and immunostaining showed increases in excitatory synaptic inputs as well as c-fos expression in Auts2 mutant brains, suggesting that an altered balance of excitatory and inhibitory inputs enhances brain excitability. Auts2 mutant mice exhibited autistic-like behaviors including impairments in social interaction and altered vocal communication. Together, these findings suggest that AUTS2 regulates excitatory synapse number to coordinate E/I balance in the brain, whose impairment may underlie the pathology of psychiatric disorders in individuals with AUTS2 mutations. AUTS2 regulates excitatory synapse number in forebrain pyramidal neurons Loss of Auts2 leads to increased spine formation in development and adulthood Loss of Auts2 alters the balance of excitatory and inhibitory synaptic inputs Auts2 mutant mice exhibit cognitive and sociobehavioral deficits
Collapse
|
83
|
Rodríguez-Berdini L, Ferrero GO, Bustos Plonka F, Cardozo Gizzi AM, Prucca CG, Quiroga S, Caputto BL. The moonlighting protein c-Fos activates lipid synthesis in neurons, an activity that is critical for cellular differentiation and cortical development. J Biol Chem 2020; 295:8808-8818. [PMID: 32385110 DOI: 10.1074/jbc.ra119.010129] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 04/28/2020] [Indexed: 01/07/2023] Open
Abstract
Differentiation of neuronal cells is crucial for the development and function of the nervous system. This process involves high rates of membrane expansion, during which the synthesis of membrane lipids must be tightly regulated. In this work, using a variety of molecular and biochemical assays and approaches, including immunofluorescence microscopy and FRET analyses, we demonstrate that the proto-oncogene c-Fos (c-Fos) activates cytoplasmic lipid synthesis in the central nervous system and thereby supports neuronal differentiation. Specifically, in hippocampal primary cultures, blocking c-Fos expression or its activity impairs neuronal differentiation. When examining its subcellular localization, we found that c-Fos co-localizes with endoplasmic reticulum markers and strongly interacts with lipid-synthesizing enzymes, whose activities were markedly increased in vitro in the presence of recombinant c-Fos. Of note, the expression of c-Fos dominant-negative variants capable of blocking its lipid synthesis-activating activity impaired neuronal differentiation. Moreover, using an in utero electroporation model, we observed that neurons with blocked c-Fos expression or lacking its AP-1-independent activity fail to initiate cortical development. These results highlight the importance of c-Fos-mediated activation of lipid synthesis for proper nervous system development.
Collapse
Affiliation(s)
- Lucia Rodríguez-Berdini
- Centro de Investigaciones en Química Biológica de Córdoba (Consejo Nacional de Investigaciones Científicas y Técnicas), Departamento de Química Biológica "Ranwel Caputto", Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Gabriel Orlando Ferrero
- Centro de Investigaciones en Química Biológica de Córdoba (Consejo Nacional de Investigaciones Científicas y Técnicas), Departamento de Química Biológica "Ranwel Caputto", Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Florentyna Bustos Plonka
- Centro de Investigaciones en Química Biológica de Córdoba (Consejo Nacional de Investigaciones Científicas y Técnicas), Departamento de Química Biológica "Ranwel Caputto", Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Andrés Mauricio Cardozo Gizzi
- Centro de Investigaciones en Química Biológica de Córdoba (Consejo Nacional de Investigaciones Científicas y Técnicas), Departamento de Química Biológica "Ranwel Caputto", Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - César Germán Prucca
- Centro de Investigaciones en Química Biológica de Córdoba (Consejo Nacional de Investigaciones Científicas y Técnicas), Departamento de Química Biológica "Ranwel Caputto", Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Santiago Quiroga
- Centro de Investigaciones en Química Biológica de Córdoba (Consejo Nacional de Investigaciones Científicas y Técnicas), Departamento de Química Biológica "Ranwel Caputto", Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Beatriz Leonor Caputto
- Centro de Investigaciones en Química Biológica de Córdoba (Consejo Nacional de Investigaciones Científicas y Técnicas), Departamento de Química Biológica "Ranwel Caputto", Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.
| |
Collapse
|
84
|
Mechanisms of action of clozapine in the treatment of neuroleptic-resistant and neuroleptic-intolerant schizophrenia. Eur Psychiatry 2020; 10 Suppl 1:39s-46s. [DOI: 10.1016/0767-399x(96)80083-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
SummaryThe mechanisms of action which account for the effectiveness of clozapine as a pharmacotherapy for the treatment of neuroleptic non-responders and neuroleptic intolerant schizophrenic subjects remain elusive. We review recent data concerning the actions of clozapine in laboratory animals, and discuss the likely sites of action of clozapine and the receptors through which clozapine acts. We suggest that actions at dopamine D2 receptors in the caudate nucleus and putamen underlie the extrapyramidal side effects of conventional neuroleptics. In contrast, we propose that clozapine acts in the prefrontal cortex, specifically targeting an as yet unidentified DA receptor of the D2 family, to exert therapeutic actions in neuroleptic non-responders. We suggest that the ability of clozapine to augment extracellular dopamine levels in the prefrontal cortex may represent a key mechanism contributing to the therapeutic effects of this drug, and suggest some alternative approaches which might be expected to result in effects similar to those of clozapine.
Collapse
|
85
|
Alachkar A, Azimullah S, Lotfy M, Adeghate E, Ojha SK, Beiram R, Łażewska D, Kieć-Kononowicz K, Sadek B. Antagonism of Histamine H3 receptors Alleviates Pentylenetetrazole-Induced Kindling and Associated Memory Deficits by Mitigating Oxidative Stress, Central Neurotransmitters, and c-Fos Protein Expression in Rats. Molecules 2020; 25:molecules25071575. [PMID: 32235506 PMCID: PMC7181068 DOI: 10.3390/molecules25071575] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 03/26/2020] [Accepted: 03/27/2020] [Indexed: 12/19/2022] Open
Abstract
Histamine H3 receptors (H3Rs) are involved in several neuropsychiatric diseases including epilepsy. Therefore, the effects of H3R antagonist E177 (5 and 10 mg/kg, intraperitoneal (i.p.)) were evaluated on the course of kindling development, kindling-induced memory deficit, oxidative stress levels (glutathione (GSH), malondialdehyde (MDA), catalase (CAT), and superoxide dismutase (SOD)), various brain neurotransmitters (histamine (HA), acetylcholine (ACh), γ-aminobutyric acid (GABA)), and glutamate (GLU), acetylcholine esterase (AChE) activity, and c-Fos protein expression in pentylenetetrazole (PTZ, 40 mg/kg) kindled rats. E177 (5 and 10 mg/kg, i.p.) significantly decreased seizure score, increased step-through latency (STL) time in inhibitory avoidance paradigm, and decreased transfer latency time (TLT) in elevated plus maze (all P < 0.05). Moreover, E177 mitigated oxidative stress by significantly increasing GSH, CAT, and SOD, and decreasing the abnormal level of MDA (all P < 0.05). Furthermore, E177 attenuated elevated levels of hippocampal AChE, GLU, and c-Fos protein expression, whereas the decreased hippocampal levels of HA and ACh were modulated in PTZ-kindled animals (all P < 0.05). The findings suggest the potential of H3R antagonist E177 as adjuvant to antiepileptic drugs with an added advantage of preventing cognitive impairment, highlighting the H3Rs as a potential target for the therapeutic management of epilepsy with accompanied memory deficits.
Collapse
Affiliation(s)
- Alaa Alachkar
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 17666, UAE; (A.A.); (S.A.); (S.K.O.); (R.B.)
| | - Sheikh Azimullah
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 17666, UAE; (A.A.); (S.A.); (S.K.O.); (R.B.)
| | - Mohamed Lotfy
- Department of Biology, College of Science, United Arab Emirates University, Al Ain 17666, UAE;
| | - Ernest Adeghate
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 17666, UAE;
| | - Shreesh K. Ojha
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 17666, UAE; (A.A.); (S.A.); (S.K.O.); (R.B.)
| | - Rami Beiram
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 17666, UAE; (A.A.); (S.A.); (S.K.O.); (R.B.)
| | - Dorota Łażewska
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Technology and Biotechnology of Drugs, Medyczna 9 St., 30-688 Kraków, Poland; (D.Ł.); (K.K.-K.)
| | - Katarzyna Kieć-Kononowicz
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Technology and Biotechnology of Drugs, Medyczna 9 St., 30-688 Kraków, Poland; (D.Ł.); (K.K.-K.)
| | - Bassem Sadek
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 17666, UAE; (A.A.); (S.A.); (S.K.O.); (R.B.)
- Correspondence: ; Tel.: +971-3-7137-512; Fax: +971-3-7672-033
| |
Collapse
|
86
|
Choi JE, Kim J, Kim J. Capturing activated neurons and synapses. Neurosci Res 2020; 152:25-34. [DOI: 10.1016/j.neures.2019.12.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/27/2019] [Accepted: 11/28/2019] [Indexed: 12/12/2022]
|
87
|
Sharma V, Kaur S. The Effect of Probiotic Intervention in Ameliorating the Altered Central Nervous System Functions in Neurological Disorders: A Review. Open Microbiol J 2020. [DOI: 10.2174/1874285802014010018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
There has been a significant rise in the occurrence of various neurological ailments worldwide. The need to investigate newer and safer intervention therapies with prophylactic and/or therapeutic effects is well understood. Probiotics have recently been shown to hold promise as an intervention option that warrants future work. Probiotic strains have shown beneficial treatment outcomes as evidenced in various animal and human studies. Although numerous articles have highlighted the role of gut microbiota and its cross-talk with human brain in modulating Central Nervous System (CNS) physiology and neurochemistry, the present review solely focuses on the ability of externally administered probiotic strains (that may or may not be part of the already existing gut microflora of an average human) in ameliorating the altered CNS functions in patients. The review aims at giving a comprehensive analysis of the studies performed on animals and humans and discusses the findings in different neurological and psychiatric disorders (Anxiety, Major Depressive disorder, bipolar disorder, schizophrenia, autism spectrum disorder, cognitive impairmentsetc). The article also highlights different mechanisms through which the probiotic bacteria operate in improving neurologic manifestations or decreasing the incidence of neurological disorders. These underlying mechanisms include both direct as well as indirect pathways involving neural, hormonal and immunological pathways. The potential of probiotics as an important dietary modification as well as a useful intervention therapy with preventive and therapeutic value for the target population holds strong. However, future evaluation into formulation designing, selecting the best probiotic strain(s) for each specific disease and safety and tolerability aspects in patients needs to be considered.
Collapse
|
88
|
Sackett DA, Moschak TM, Carelli RM. Nucleus accumbens shell dopamine mediates outcome value, but not predicted value, in a magnitude decision-making task. Eur J Neurosci 2020; 51:1526-1538. [PMID: 31863510 DOI: 10.1111/ejn.14655] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 11/06/2019] [Accepted: 12/12/2019] [Indexed: 11/29/2022]
Abstract
Effective decision-making depends on an animal's ability to predict and select the outcome of greatest value, and the nucleus accumbens (NAc) and its dopaminergic input play a key role in this process. We previously reported that rapid dopamine release in the NAc shell preferentially tracks the "preferred" (i.e., large reward) option during cues that predict the ability to respond for rewards of different sizes, as well as during reward delivery itself. The present study assessed whether shell dopamine release at these discrete times selectively mediated choice behavior for rewards of different magnitudes using optogenetics. Here, using Long Evans TH:Cre± rats we employed selective optogenetic stimulation of dopamine terminals in the NAc shell during either reward-predictive cues (experiment 1) or reward delivery (experiment 2) in a magnitude-based decision-making task. We found that in TH:Cre± rats, but not littermate controls, optical stimulation during low-magnitude reward delivery during forced choice trials was sufficient to bias preference for this option when given a choice. In contrast, optical stimulation of shell dopamine terminals during low-magnitude reward-predictive cues in forced choice trials did not shift free choice behavior in TH:Cre± rats or controls. The findings indicate that preferential dopamine signaling in the NAc shell during reward outcome (delivery), but not reward-predictive cues are sufficient to influence choice behavior in our task supporting a causal role of dopamine in the NAc shell in reward outcome value, but not value-based predictive strategies.
Collapse
Affiliation(s)
- Deirdre A Sackett
- Department of Psychology and Neuroscience, The University of North Carolina, Chapel Hill, NC, USA
| | - Travis M Moschak
- Department of Psychology and Neuroscience, The University of North Carolina, Chapel Hill, NC, USA
| | - Regina M Carelli
- Department of Psychology and Neuroscience, The University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
89
|
Nishimura H, Kawasaki M, Matsuura T, Suzuki H, Motojima Y, Baba K, Ohnishi H, Yamanaka Y, Fujitani T, Yoshimura M, Maruyama T, Ueno H, Sonoda S, Nishimura K, Tanaka K, Sanada K, Onaka T, Ueta Y, Sakai A. Acute Mono-Arthritis Activates the Neurohypophysial System and Hypothalamo-Pituitary Adrenal Axis in Rats. Front Endocrinol (Lausanne) 2020; 11:43. [PMID: 32117068 PMCID: PMC7026388 DOI: 10.3389/fendo.2020.00043] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 01/23/2020] [Indexed: 01/04/2023] Open
Abstract
Various types of acute/chronic nociceptive stimuli cause neuroendocrine responses such as activation of the hypothalamo-neurohypophysial [oxytocin (OXT) and arginine vasopressin (AVP)] system and hypothalamo-pituitary adrenal (HPA) axis. Chronic multiple-arthritis activates the OXT/AVP system, but the effects of acute mono-arthritis on the OXT/AVP system in the same animals has not been simultaneously evaluated. Further, AVP, not corticotropin-releasing hormone (CRH), predominantly activates the HPA axis in chronic multiple-arthritis, but the participation of AVP in HPA axis activation in acute mono-arthritis remains unknown. Therefore, we aimed to simultaneously evaluate the effects of acute mono-arthritis on the activity of the OXT/AVP system and the HPA axis. In the present study, we used an acute mono-arthritic model induced by intra-articular injection of carrageenan in a single knee joint of adult male Wistar rats. Acute mono-arthritis was confirmed by a significant increase in knee diameter in the carrageenan-injected knee and a significant decrease in the mechanical nociceptive threshold in the ipsilateral hind paw. Immunohistochemical analysis revealed that the number of Fos-immunoreactive (ir) cells in the ipsilateral lamina I-II of the dorsal horn was significantly increased, and the percentage of OXT-ir and AVP-ir neurons expressing Fos-ir in both sides of the supraoptic (SON) and paraventricular nuclei (PVN) was increased in acute mono-arthritic rats. in situ hybridization histochemistry revealed that levels of OXT mRNA and AVP hnRNA in the SON and PVN, CRH mRNA in the PVN, and proopiomelanocortin mRNA in the anterior pituitary were also significantly increased in acute mono-arthritic rats. Further, plasma OXT, AVP, and corticosterone levels were significantly increased in acute mono-arthritic rats. These results suggest that acute mono-arthritis activates ipsilateral nociceptive afferent pathways at the spinal level and causes simultaneous and integrative activation of the OXT/AVP system. In addition, the HPA axis is activated by both AVP and CRH in acute mono-arthritis with a distinct pattern compared to that in chronic multiple-arthritis.
Collapse
Affiliation(s)
- Haruki Nishimura
- Department of Orthopaedics Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Makoto Kawasaki
- Department of Orthopaedics Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
- *Correspondence: Makoto Kawasaki
| | - Takanori Matsuura
- Department of Orthopaedics Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Hitoshi Suzuki
- Department of Orthopaedics Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yasuhito Motojima
- Department of Orthopaedics Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Kazuhiko Baba
- Department of Orthopaedics Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Hideo Ohnishi
- Department of Orthopaedics Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yoshiaki Yamanaka
- Department of Orthopaedics Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Teruaki Fujitani
- Department of Orthopaedics Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Mitsuhiro Yoshimura
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Takashi Maruyama
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Hiromichi Ueno
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Satomi Sonoda
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Kazuaki Nishimura
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Kentarou Tanaka
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Kenya Sanada
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Tatsushi Onaka
- Division of Brain and Neurophysiology, Department of Physiology, Jichi Medical University, Shimotsuke, Japan
| | - Yoichi Ueta
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Akinori Sakai
- Department of Orthopaedics Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| |
Collapse
|
90
|
Nguyen T, Wen S, Gong M, Yuan X, Xu D, Wang C, Jin J, Zhou L. Dapagliflozin Activates Neurons in the Central Nervous System and Regulates Cardiovascular Activity by Inhibiting SGLT-2 in Mice. Diabetes Metab Syndr Obes 2020; 13:2781-2799. [PMID: 32848437 PMCID: PMC7425107 DOI: 10.2147/dmso.s258593] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 07/10/2020] [Indexed: 02/06/2023] Open
Abstract
PURPOSE This study investigates the possible effect and central mechanism of novel antidiabetic medication sodium glucose transporter-2 (SGLT-2i) on the cardiovascular activity. MATERIAL AND METHODS Thirty-four normal male C57BL/6 mice were randomly assigned to 2 groups to receive single Dapagliflozin (1.52mg/kg) dose via intragastric gavage or a comparable dose of saline. Glycemic level (BG), blood pressure (BP) and heart rate (HR) were measured 2 hours after administration of the respective treatments. Immunohistochemical tests were performed to determine the effect of SGLT-2i on neural localization of SGLT-2 and c-Fos, a neural activator. The distributional relationships of SGLT-2 and c-Fos were examined by immunofluorescence. RESULTS Administration of SGLT-2i significantly decreased BP but did not affect the HR. There was no difference in BG between the two groups. Results showed that SGLT-2 was localized to specific regions involved in autonomic control. Expression of c-Fos was significantly higher in major critical nuclei in the aforementioned regions in groups treated with Dapagliflozin. CONCLUSION This study demonstrates that SGLT-2 is expressed in CNS tissues involved in autonomic control and possibly influence cardiovascular function. Dapagliflozin influences central autonomic activity via unidentified pathways by inhibiting central or peripheral SGLT-2. These results provide a new concept that sympathetic inhibition by SGLT-2i can be mediated by central autonomic system, a mechanism that explains how SGLT-2i improves the cardiovascular function.
Collapse
Affiliation(s)
- Thiquynhnga Nguyen
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai201399, People’s Republic of China
| | - Song Wen
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai201399, People’s Republic of China
| | - Min Gong
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai201399, People’s Republic of China
| | - Xinlu Yuan
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai201399, People’s Republic of China
| | - Dongxiang Xu
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai201399, People’s Republic of China
| | - Chaoxun Wang
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai201399, People’s Republic of China
| | - Jianlan Jin
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai201399, People’s Republic of China
| | - Ligang Zhou
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai201399, People’s Republic of China
- Correspondence: Ligang Zhou Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai201399, People’s Republic of ChinaTel +86 13611927616 Email
| |
Collapse
|
91
|
Stern SA, Doerig KR, Azevedo EP, Stoffel E, Friedman JM. Control of non-homeostatic feeding in sated mice using associative learning of contextual food cues. Mol Psychiatry 2020; 25:666-679. [PMID: 29875477 PMCID: PMC6281813 DOI: 10.1038/s41380-018-0072-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 01/31/2018] [Accepted: 03/13/2018] [Indexed: 02/07/2023]
Abstract
Feeding is a complex motivated behavior controlled by a distributed neural network that processes sensory information to generate adaptive behavioral responses. Accordingly, studies using appetitive Pavlovian conditioning confirm that environmental cues that are associated with food availability can induce feeding even in satiated subjects. However, in mice, appetitive conditioning generally requires intensive training and thus can impede molecular studies that often require large numbers of animals. To address this, we developed and validated a simple and rapid context-induced feeding (Ctx-IF) task in which cues associated with food availability can later lead to increased food consumption in sated mice. We show that the associated increase in food consumption is driven by both positive and negative reinforcement and that spaced training is more effective than massed training. Ctx-IF can be completed in ~1 week and provides an opportunity to study the molecular mechanisms and circuitry underlying non-homeostatic eating. We have used this paradigm to map brain regions that are activated during Ctx-IF with cFos immunohistochemistry and found that the insular cortex, and other regions, are activated following exposure to cues denoting the availability of food. Finally, we show that inhibition of the insular cortex using GABA agonists impairs performance of the task. Our findings provide a novel assay in mice for defining the functional neuroanatomy of appetitive conditioning and identify specific brain regions that are activated during the development of learned behaviors that impact food consumption.
Collapse
Affiliation(s)
- Sarah A. Stern
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular Genetics, Rockefeller University, New York, NY 10065 USA
| | - Katherine R. Doerig
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular Genetics, Rockefeller University, New York, NY 10065 USA
| | - Estefania P. Azevedo
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular Genetics, Rockefeller University, New York, NY 10065 USA
| | - Elina Stoffel
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular Genetics, Rockefeller University, New York, NY 10065 USA
| | - Jeffrey M. Friedman
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular Genetics, Rockefeller University, New York, NY 10065 USA ,0000 0001 2167 1581grid.413575.1Howard Hughes Medical Institute, Chevy Chase, MD USA
| |
Collapse
|
92
|
Pinna A, Costa G, Contu L, Morelli M. Fos expression induced by olanzapine and risperidone in the central extended amygdala. Eur J Pharmacol 2019; 865:172764. [PMID: 31678081 DOI: 10.1016/j.ejphar.2019.172764] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 10/04/2019] [Accepted: 10/29/2019] [Indexed: 12/12/2022]
Abstract
The extended amygdala has been proposed to play an essential role in cognitive and affective processes and in neuropsychiatric disorders. In the present study, we examined the induction of Fos-like nuclei in the central amygdaloid nucleus (CeA), sublenticular extended amygdala (SLEA), interstitial nucleus of the posterior limb of the anterior commissure (IPAC), and bed nucleus of the stria terminalis (BSTL) of rodents to improve the knowledge regarding the pharmacological profile, therapeutic efficacy, and side-effects of olanzapine, an atypical antipsychotic drug and risperidone, a mixed atypical/typical antipsychotic drug in the rat brain. In addition, we evaluated the induction of Fos-like-nuclei in areas connected with these structures such as prefrontal cortex (PFCx), and nucleus accumbens shell, and in other important areas including the lateral septum and caudate-putamen that are involved in the therapeutic efficacy or side-effects of antipsychotic drugs. Fos-like-immunoreactivity induced by olanzapine and risperidone was compared with that by the atypical antipsychotic clozapine and typical antipsychotic haloperidol. Regarding the extended amygdala, and similarly to clozapine, olanzapine (5-10 mg/kg) and, with a lower efficacy, risperidone (1-3 mg/kg), induced Fos-like-nuclei in CeA, IPAC, SLEA, and BSTL. Both these drugs increased the induction of Fos-like-nuclei in PFCx, nucleus accumbens shell, lateral septum, and caudate-putamen. On the contrary, the increase of Fos-like-nuclei in the extended amygdala by haloperidol was restricted to IPAC only. These findings, consistent with the important role of extended amygdala in neuropsychiatric disorders characterized by affective disturbances, showed that olanzapine and risperidone, contrary to haloperidol, preferentially activated Fos-expression in these brain areas.
Collapse
Affiliation(s)
- Annalisa Pinna
- National Research Council of Italy, Neuroscience Institute - Cagliari, Cittadella Universitaria, Blocco A, SP 8, Km 0.700, 09042, Monserrato, CA, Italy.
| | - Giulia Costa
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cittadella Universitaria, Blocco A, SP 8, Km 0.700, 09042, Monserrato, CA, Italy.
| | - Liliana Contu
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cittadella Universitaria, Blocco A, SP 8, Km 0.700, 09042, Monserrato, CA, Italy.
| | - Micaela Morelli
- National Research Council of Italy, Neuroscience Institute - Cagliari, Cittadella Universitaria, Blocco A, SP 8, Km 0.700, 09042, Monserrato, CA, Italy; Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cittadella Universitaria, Blocco A, SP 8, Km 0.700, 09042, Monserrato, CA, Italy.
| |
Collapse
|
93
|
Ni RJ, Wang J, Shu YM, Xu L, Zhou JN. Mapping of c-Fos expression in male tree shrew forebrain. Neurosci Lett 2019; 714:134603. [PMID: 31693931 DOI: 10.1016/j.neulet.2019.134603] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 10/28/2019] [Accepted: 10/29/2019] [Indexed: 02/05/2023]
Abstract
The tree shrew is susceptible to stimuli. However, mapping of c-Fos expression in male tree shrew forebrain has not been explored. The present results provided the first detailed mapping of c-Fos expression in the forebrain of the tree shrew (Tupaia belangeri chinensis). Acute restraint stress rapidly increased the density of c-Fos-immunoreactive (-ir) neurons in the medial orbital cortex (MO), infralimbic cortex, intermediate part of the lateral septal nucleus (LSi), ventral part of the lateral septal nucleus (LSv), anterior part of the bed nucleus of the stria terminalis, posterior part of the bed nucleus of the stria terminalis (STP), paraventricular nucleus of the hypothalamus, supraoptic nucleus, lateral hypothalamic area, ventromedial hypothalamic nucleus (VMH), and medial amygdaloid nucleus (MeA). Furthermore, a significant increase in c-Fos expression was observed in the MO, LSi, LSv, STP, VMH, arcuate hypothalamic nucleus, anterior amygdaloid area, MeA, and cortical amygdaloid nucleus immediately after acute footshock stress. In addition, the distinct patterns of c-Fos expression in the forebrain were shown in context-, restraint-, or footshock-treated tree shrews. In general, the present study provides the first detailed maps of c-Fos expression in male tree shrew forebrain immediately after various stimuli.
Collapse
Affiliation(s)
- Rong-Jun Ni
- Psychiatric Laboratory and Mental Health Center, West China Hospital of Sichuan University, Chengdu 610041, China; Chinese Academy of Science Key Laboratory of Brain Function and Diseases, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China; Huaxi Brain Research Center, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Jing Wang
- Department of Neurobiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Key Laboratory of Animal Models and Human Disease Mechanisms, and Laboratory of Learning and Memory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Yu-Mian Shu
- School of Architecture and Civil Engineering, Chengdu University, Chengdu 610041, China
| | - Lin Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms, and Laboratory of Learning and Memory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Jiang-Ning Zhou
- Chinese Academy of Science Key Laboratory of Brain Function and Diseases, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China.
| |
Collapse
|
94
|
Kovács LÁ, Berta G, Csernus V, Ujvári B, Füredi N, Gaszner B. Corticotropin-Releasing Factor-Producing Cells in the Paraventricular Nucleus of the Hypothalamus and Extended Amygdala Show Age-Dependent FOS and FOSB/DeltaFOSB Immunoreactivity in Acute and Chronic Stress Models in the Rat. Front Aging Neurosci 2019; 11:274. [PMID: 31649527 PMCID: PMC6794369 DOI: 10.3389/fnagi.2019.00274] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 09/24/2019] [Indexed: 01/01/2023] Open
Abstract
Corticotropin-releasing factor (CRF) immunoreactive (ir) neurons of the paraventricular nucleus of the hypothalamus (PVN) play pivotal role in the coordination of stress response. CRF-producing cells in the central nucleus of amygdala (CeA) and oval division of the bed nucleus of stria terminalis (BNSTov) are also involved in stress adaptation and mood control. Immediate early gene products, subunits of the transcription factor activator protein 1 (AP1) are commonly used as acute (FOS) and/or chronic (FOSB/deltaFOSB) markers for the neuronal activity in stress research. It is well known that the course of aging affects stress adaptation, but little is known about the aging-related stress sensitivity of CRF neurons. To the best of our knowledge, the stress-induced neuronal activity of CRF neurons in the course of aging in acute and chronic stress models was not studied systematically yet. Therefore, the aim of the present study was to quantify the acute restraint stress (ARS) and chronic variable mild stress (CVMS) evoked neuronal activity in CRF cells of the PVN, CeA, and BNSTov using triple-label immunofluorescence throughout the whole lifespan in the rat. We hypothesized that the FOS and FOSB content of CRF cells upon ARS or CVMS decreases with age. Our results showed that the FOS and FOSB response to ARS declined with age in the PVN-CRF cells. BNSTov and CeA CRF cells did not show remarkable stress-induced elevation of these markers neither in ARS, nor in CVMS. Exposure to CVMS resulted in an age-independent significant increase of FOSB/delta FOSB immunosignal in PVN-CRF neurons. Unexpectedly, we detected a remarkable stress-independent FOSB/deltaFOSB signal in CeA- and BNSTov-CRF cells that declined with the course of aging. In summary, PVN-CRF cells show decreasing acute stress sensitivity (i.e., FOS and FOSB immunoreactivity) with the course of aging, while their (FOSB/deltaFOSB) responsivity to chronic challenge is maintained till senescence. Stress exposure does not affect the occurrence of the examined Fos gene products in CeA- and BNSTov-CRF cells remarkably suggesting that their contribution to stress adaptation response does not require AP1-controlled transcriptional changes.
Collapse
Affiliation(s)
- László Á Kovács
- Department of Anatomy, University of Pécs Medical School, Pécs, Hungary.,Centre for Neuroscience, Pécs University, Pécs, Hungary
| | - Gergely Berta
- Department of Medical Biology and Central Electron Microscope Laboratory, University of Pécs Medical School, Pécs, Hungary
| | - Valér Csernus
- Department of Anatomy, University of Pécs Medical School, Pécs, Hungary
| | - Balázs Ujvári
- Department of Anatomy, University of Pécs Medical School, Pécs, Hungary.,Centre for Neuroscience, Pécs University, Pécs, Hungary
| | - Nóra Füredi
- Department of Anatomy, University of Pécs Medical School, Pécs, Hungary.,Centre for Neuroscience, Pécs University, Pécs, Hungary
| | - Balázs Gaszner
- Department of Anatomy, University of Pécs Medical School, Pécs, Hungary.,Centre for Neuroscience, Pécs University, Pécs, Hungary
| |
Collapse
|
95
|
Zhou M, Liu Y, He Y, Xie K, Quan D, Tang Y, Huang H, Huang C. Selective chemical ablation of transient receptor potential vanilloid 1 expressing neurons in the left stellate ganglion protects against ischemia-induced ventricular arrhythmias in dogs. Biomed Pharmacother 2019; 120:109500. [PMID: 31600641 DOI: 10.1016/j.biopha.2019.109500] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 09/12/2019] [Accepted: 09/26/2019] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVES Findings from prior investigations show that left stellate ganglion (LSG) inhibitory approaches protect the heart from ventricular arrhythmias (VAs) caused by acute myocardial infarction (AMI), which still remain many side effects. Targeted transient receptor potential vanilloid 1/tyrosine hydroxylase (TRPV-1/TH) expressing sympathetic neurons ablation is a novel neuro-ablative strategy. The aim of this investigation was to explore if targeted molecular neuro-ablative strategy by resiniferatoxin (RTX) stellate microinjection could protect against ischemia-induced VAs. METHODS Twenty-four anesthetized beagles were assigned to a control group (n = 12) and RTX group (n = 12) in a random manner. Targeted molecular neuro-ablative was produced by RTX stellate microinjection and DMSO was microinjected into LSG in the same way as control. Plasma norepinephrine (NE) level, heart rate variability (HRV), Tpeak-Tend interval (Tp-Te), LSG neural activity and function, ventricular effective refractory period (ERP), beat-to-beat variability of repolarization (BVR) and ventricular action potential duration (APD) were measured at baseline and 60 min after RTX or DMSO microinjection. AMI model was established by the ligation of left anterior descending coronary artery and 60-minute electrocardiography was continuously recorded for VAs analysis. Subsequently, HRV, Tp-Te, plasma NE level from jugular vein and coronary sinus, LSG neural activity and function, ventricular ERP, ventricular APD, BVR, action potential duration alternans (APDA) cycle length and ventricular fibrillation threshold (VFT) were evaluated after AMI. Finally, tissue collection of LSG was performed for examining the TRPV-1, nerve growth factor (NGF) protein and c-fos protein. RESULTS TRPV-1 was highly expressed in the TH-expressing neurons and RTX injection significantly ablated TRPV-1/TH-positive neurons in LSG. Compared with baseline, RTX stellate microinjection significantly reduced plasma NE level, the sympathetic component of HRV, LSG neural activity and LSG function, shortened Tp-Te, prolonged ventricular ERP and APD, but there were no remarkable differences existed for control group. AMI resulted in the significant raise in plasma NE level from jugular vein and coronary sinus, the sympathetic component of HRV, LSG neural activity and LSG function, the marked prolongation in Tp-Te and BVR, the significant decrease in ERP and APD from ischemia area, and the increase in APDA cycle length in the ischemic region of the control group, which were remarkably attenuated in the RTX group. RTX pretreatment markedly rose the VFT in the RTX group. Furthermore, the AMI-triggered VAs was significantly prevented by RTX injection in the RTX group. RTX microinjection down-regulated significantly TRPV-1, NGF and c-fos expression in the LSG compared with the control group. CONCLUSION Targeted ablation of TRPV-1/TH positive sympathetic neurons induced by RTX stellate microinjection could suppress ischemia-induced cardiac autonomic imbalances and cardiac electrophysiology instability to protect against AMI-induced VAs.
Collapse
Affiliation(s)
- Mingmin Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute of Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yu Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute of Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China.
| | - Yan He
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute of Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Ke Xie
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute of Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Dajun Quan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute of Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yanhong Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute of Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - He Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute of Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Congxin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute of Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
96
|
Gastrin releasing peptide-induced satiety is associated with hypothalamic and brainstem changes in chicks. Neurosci Lett 2019; 713:134529. [PMID: 31585210 DOI: 10.1016/j.neulet.2019.134529] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 09/24/2019] [Accepted: 09/30/2019] [Indexed: 11/23/2022]
Abstract
Gastrin releasing peptide (GRP) is involved in the stimulation of gastric acid release from the stomach. It also mediates effects on feeding behavior. It is associated with anorexigenic effects in both mammalian and avian species, but the mechanism of action is unknown in any species. The aim of the present study was thus to investigate the hypothalamic and brainstem mechanisms mediating GRP-induced satiety in chicks. In Experiment 1, chicks that received intracerebroventricular (ICV) injection of GRP reduced food intake for up to 150 min following injection and reduced water intake up to 120 min following injection. In Experiment 2, chicks that were food restricted following GRP injection did not reduce water intake. Alimentary canal transit time was not affected by GRP in Experiment 3. A behavior analysis was conducted in Experiment 4, revealing that GRP-treated chicks reduced feeding pecks. In Experiment 5, GRP-treated chicks had increased c-Fos immunoreactivity in the lateral hypothalamus, paraventricular nucleus, and arcuate nucleus of the hypothalamus, and the nucleus of the solitary tract. Collectively, these results demonstrate that central GRP causes anorexigenic effects that are associated with hypothalamic changes without affecting other behaviors.
Collapse
|
97
|
Rao S, Chen R, LaRocca AA, Christiansen MG, Senko AW, Shi CH, Chiang PH, Varnavides G, Xue J, Zhou Y, Park S, Ding R, Moon J, Feng G, Anikeeva P. Remotely controlled chemomagnetic modulation of targeted neural circuits. NATURE NANOTECHNOLOGY 2019; 14:967-973. [PMID: 31427746 PMCID: PMC6778020 DOI: 10.1038/s41565-019-0521-z] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 07/03/2019] [Indexed: 05/20/2023]
Abstract
Connecting neural circuit output to behaviour can be facilitated by the precise chemical manipulation of specific cell populations1,2. Engineered receptors exclusively activated by designer small molecules enable manipulation of specific neural pathways3,4. However, their application to studies of behaviour has thus far been hampered by a trade-off between the low temporal resolution of systemic injection versus the invasiveness of implanted cannulae or infusion pumps2. Here, we developed a remotely controlled chemomagnetic modulation-a nanomaterials-based technique that permits the pharmacological interrogation of targeted neural populations in freely moving subjects. The heat dissipated by magnetic nanoparticles (MNPs) in the presence of alternating magnetic fields (AMFs) triggers small-molecule release from thermally sensitive lipid vesicles with a 20 s latency. Coupled with the chemogenetic activation of engineered receptors, this technique permits the control of specific neurons with temporal and spatial precision. The delivery of chemomagnetic particles to the ventral tegmental area (VTA) allows the remote modulation of motivated behaviour in mice. Furthermore, this chemomagnetic approach activates endogenous circuits by enabling the regulated release of receptor ligands. Applied to an endogenous dopamine receptor D1 (DRD1) agonist in the nucleus accumbens (NAc), a brain area involved in mediating social interactions, chemomagnetic modulation increases sociability in mice. By offering a temporally precise control of specified ligand-receptor interactions in neurons, this approach may facilitate molecular neuroscience studies in behaving organisms.
Collapse
Affiliation(s)
- Siyuan Rao
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Simons Center for Social Brain, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ritchie Chen
- Simons Center for Social Brain, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Ava A LaRocca
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Michael G Christiansen
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Health Sciences and Technology at the Swiss Federal Institute of Technology in Zürich (ETHZ), Zürich, Switzerland
| | - Alexander W Senko
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Cindy H Shi
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Po-Han Chiang
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Georgios Varnavides
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Jian Xue
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yang Zhou
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Seongjun Park
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ruihua Ding
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Junsang Moon
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Guoping Feng
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Polina Anikeeva
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
98
|
Dedic N, Kühne C, Gomes KS, Hartmann J, Ressler KJ, Schmidt MV, Deussing JM. Deletion of CRH From GABAergic Forebrain Neurons Promotes Stress Resilience and Dampens Stress-Induced Changes in Neuronal Activity. Front Neurosci 2019; 13:986. [PMID: 31619956 PMCID: PMC6763571 DOI: 10.3389/fnins.2019.00986] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 09/02/2019] [Indexed: 12/15/2022] Open
Abstract
Dysregulation of the corticotropin-releasing hormone (CRH) system has been implicated in stress-related psychopathologies such as depression and anxiety. Although most studies have linked CRH/CRH receptor 1 signaling to aversive, stress-like behavior, recent work has revealed a crucial role for distinct CRH circuits in maintaining positive emotional valence and appetitive responses under baseline conditions. Here we addressed whether deletion of CRH, specifically from GABAergic forebrain neurons (Crh CKO-GABA mice) differentially affects general behavior under baseline and chronic stress conditions. Expression mapping in Crh CK O-GABA mice revealed absence of Crh in GABAergic neurons of the cortex and limbic regions including the hippocampus, central nucleus of the amygdala and the bed nucleus of the stria terminals, but not in the paraventricular nucleus of hypothalamus. Consequently, conditional CRH knockout animals exhibited no alterations in circadian and stress-induced corticosterone release compared to controls. Under baseline conditions, absence of Crh from forebrain GABAergic neurons resulted in social interaction deficits but had no effect on other behavioral measures including locomotion, anxiety, immobility in the forced swim test, acoustic startle response and fear conditioning. Interestingly, following exposure to chronic social defeat stress, Crh CKO-GABA mice displayed a resilient phenotype, which was accompanied by a dampened, stress-induced expression of immediate early genes c-fos and zif268 in several brain regions. Collectively our data reveals the requirement of GABAergic CRH circuits in maintaining appropriate social behavior in naïve animals and further supports the ability of CRH to promote divergent behavioral states under baseline and severe stress conditions.
Collapse
Affiliation(s)
- Nina Dedic
- Molecular Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany.,Department of Psychiatry, Harvard Medical School and McLean Hospital, Belmont, MA, United States
| | - Claudia Kühne
- Molecular Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Karina S Gomes
- Molecular Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany.,Laboratory of Neuropsychopharmacology, Paulista State University, Araraquara, Brazil
| | - Jakob Hartmann
- Department of Psychiatry, Harvard Medical School and McLean Hospital, Belmont, MA, United States.,Stress Resilience, Max Planck Institute of Psychiatry, Munich, Germany
| | - Kerry J Ressler
- Department of Psychiatry, Harvard Medical School and McLean Hospital, Belmont, MA, United States
| | - Mathias V Schmidt
- Stress Resilience, Max Planck Institute of Psychiatry, Munich, Germany
| | - Jan M Deussing
- Molecular Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| |
Collapse
|
99
|
Wall KD, Olivos DR, Rinaman L. High Fat Diet Attenuates Cholecystokinin-Induced cFos Activation of Prolactin-Releasing Peptide-Expressing A2 Noradrenergic Neurons in the Caudal Nucleus of the Solitary Tract. Neuroscience 2019; 447:113-121. [PMID: 31518655 DOI: 10.1016/j.neuroscience.2019.08.054] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 08/22/2019] [Accepted: 08/28/2019] [Indexed: 10/26/2022]
Abstract
Cholecystokinin (CCK) released from the small intestine increases the activity of vagal afferents that relay satiety signals to the caudal nucleus of the solitary tract (cNTS). A caudal subset of A2 noradrenergic neurons within the cNTS that express prolactin-releasing peptide (PrRP) have been proposed to mediate CCK-induced satiety. However, the ability of exogenous CCK to activate cFos expression by PrRP neurons has only been reported in rats and mice after a very high dose (i.e., 50 μg/kg BW) that also activates the hypothalamic-pituitary-adrenal stress axis. The present study examined the ability of a much lower CCK dose (1.0 µg/kg BW, i.p) to activate PrRP-positive neurons in the rat cNTS. We further examined whether maintenance of rats on high fat diet (HFD; 45% kcal from fat) alters CCK-induced activation of PrRP neurons, since HFD blunts the ability of CCK to suppress food intake. Rats maintained on HFD for 7 weeks consumed more kcal and gained more BW compared to rats maintained on Purina chow (13.5% kcal from fat). CCK-treated rats displayed increased numbers of cFos-positive cNTS neurons compared to non-injected and saline-injected controls, with no effect of diet. In chow-fed rats, a significantly larger proportion of PrRP neurons were activated after CCK treatment compared to controls; conversely, CCK did not increase PrRP neuronal activation in HFD-fed rats. Collectively, these results indicate that a relatively low dose of exogenous CCK is sufficient to activate PrRP neurons in chow-fed rats, and that this effect is blunted in rats maintained for several weeks on HFD.
Collapse
Affiliation(s)
- Kaylee D Wall
- Florida State University, Department of Psychology and Program in Neuroscience, Tallahassee, FL, USA
| | - Diana R Olivos
- University of Pittsburgh, Department of Neuroscience, Dietrich School of Arts and Sciences, Pittsburgh, PA, USA
| | - Linda Rinaman
- Florida State University, Department of Psychology and Program in Neuroscience, Tallahassee, FL, USA.
| |
Collapse
|
100
|
Pose S, Zuluaga MJ, Ferreño M, Agrati D, Bedó G, Uriarte N. Raising overlapping litters: Differential activation of rat maternal neural circuitry after interacting with newborn or juvenile pups. J Neuroendocrinol 2019; 31:e12701. [PMID: 30784145 DOI: 10.1111/jne.12701] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 02/16/2019] [Accepted: 02/18/2019] [Indexed: 01/05/2023]
Abstract
The maternal behaviour of a rat dynamically changes during the postpartum period, adjusting to the characteristics and physiological needs of the pups. This adaptation has been attributed to functional modifications in the maternal circuitry. Maternal behaviour can also flexibly adapt according to different litter compositions. Thus, mothers with two overlapping litters can concurrently take care of neonate and juvenile pups, mostly directing their attention to the newborns. We hypothesised that the maternal circuitry of these mothers would show a differential activation pattern after interacting with pups depending on the developmental stage of their offspring. Thus, we evaluated the activation of several areas of the maternal circuitry in mothers of overlapping litters, using c-Fos immunoreactivity as a marker of neuronal activation, after interacting with newborns or juveniles. The results showed that mothers with overlapping litters display different behavioural responses towards their newborn and their juvenile pups. Interestingly, these behavioural displays co-occurred with specific patterns of activation of the maternal neural circuitry. Thus, a similar expression of c-Fos was observed in some key brain areas of mothers that interacted with newborns or juveniles, such as the medial preoptic area and the nucleus accumbens, whereas a differential activation was quantified in the ventral region of the bed nucleus of the stria terminalis, the infralimbic and prelimbic subregions of the medial prefrontal cortex and the basolateral and medial nuclei of the amygdala. We posit that the specific profile of activation of the neural circuitry controlling maternal behaviour in mothers with overlapping litters enables dams to respond adequately to the newborn and the juvenile pups.
Collapse
Affiliation(s)
- Sabrina Pose
- Laboratorio de Neurociencias, Sección Biomatemática, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - María José Zuluaga
- PDU Biofisicoquímica, Centro Universitario Regional Norte - Sede Salto, Universidad de la República, Montevideo, Uruguay
| | - Marcela Ferreño
- Laboratorio de Neurociencias, Sección Biomatemática, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Daniella Agrati
- Sección Fisiología y Nutrición, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Gabriela Bedó
- Sección Genética Evolutiva, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Natalia Uriarte
- Laboratorio de Neurociencias, Sección Biomatemática, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|