1
|
McGregor R, Wu MF, Thannickal TC, Li S, Siegel JM. Opioid-induced neuroanatomical, microglial and behavioral changes are blocked by suvorexant without diminishing opioid analgesia. NATURE. MENTAL HEALTH 2024; 2:1018-1031. [PMID: 39989723 PMCID: PMC11845277 DOI: 10.1038/s44220-024-00278-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 05/31/2024] [Indexed: 02/25/2025]
Abstract
Heroin use disorder in humans and chronic opioid administration to mice result in an increase in the number and a decrease in the size of detected hypocretin (Hcrt, or orexin) neurons. Chronic morphine administration to mice increases Hcrt axonal projections to the ventral tegmental area (VTA), the level of tyrosine hydroxylase (TH) in VTA and the number of detected TH+ cells in VTA, and activates VTA and hypothalamic microglia. Co-administration of morphine with the dual Hcrt receptor antagonist suvorexant prevents morphine-induced changes in the number and size of Hcrt neurons, the increase in Hcrt projections to the VTA and microglial activation in the VTA and hypothalamus. Co-administration of suvorexant with morphine also prevents morphine anticipatory behavior and reduces opioid withdrawal symptoms. However, suvorexant does not diminish morphine analgesia. Here we show that combined administration of opioids and suvorexant may reduce the addiction potential of opioid use for pain relief in humans while maintaining the analgesic effects of opioids.
Collapse
Affiliation(s)
- Ronald McGregor
- Neuropsychiatric Institute and Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
- These authors contributed equally: Ronald McGregor, Ming-Fung Wu
| | - Ming-Fung Wu
- Neuropsychiatric Institute and Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
- These authors contributed equally: Ronald McGregor, Ming-Fung Wu
| | - Thomas C. Thannickal
- Neuropsychiatric Institute and Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Songlin Li
- Neuropsychiatric Institute and Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Jerome M. Siegel
- Neuropsychiatric Institute and Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| |
Collapse
|
2
|
Uzcategui NL, Güçer S, Richter C, Speidel A, Zirdum E, Duszenko M, Garaschuk O, Figarella K. Live imaging of microglia during sleeping sickness reveals early and heterogeneous inflammatory responses. Front Immunol 2023; 14:1253648. [PMID: 37781403 PMCID: PMC10534015 DOI: 10.3389/fimmu.2023.1253648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 08/30/2023] [Indexed: 10/03/2023] Open
Abstract
Introduction Invasion of the central nervous system (CNS) is the most serious consequence of Trypanosoma brucei infection, which causes sleeping sickness. Recent experimental data have revealed some more insights into the disease during the meningoencephalitic stage. However, detailed cellular processes befalling the CNS during the disease are poorly understood. Methods To further address this issue, we implanted a cranial window on the cortex of B6.129P2(Cg)-Cx3cr1tm1Litt/J mice, infected them with Trypanosoma brucei expressing RFP via intraperitoneal injection, and monitored microglial cells and parasites longitudinally over 30 days using in vivo 2-photon imaging. We correlated the observed changes with histological analyses to evaluate the recruitment of peripheral immune cells. Results and discussion We uncovered an early involvement of microglia that precedes invasion of the CNS by the parasite. We accomplished a detailed characterization of the progressive sequence of events that correlates with microglial morphological changes and microgliosis. Our findings unveiled a heterogeneous microglial response in places of initial homeostatic disruption near brain barriers and pointed out an exceptional capability of microglia to hamper parasite proliferation inside the brain. We also found early signs of inflammation in the meninges, which synchronize with the microglial response. Moreover, we observed a massive infiltration of peripheral immune cells into the parenchyma as a signature in the final disease stage. Overall, our study provides new insights into the host-pathogen immune interactions in the meningeal and parenchymal compartments of the neocortex.
Collapse
Affiliation(s)
- Nestor L. Uzcategui
- Department of Neurophysiology, Institute of Physiology, University of Tuebingen, Tuebingen, Germany
- Institute of Tropical Medicine, University of Tuebingen, Tuebingen, Germany
- Institute of Anatomy, Central University of Venezuela, Caracas, Venezuela
| | - Sena Güçer
- Department of Neurophysiology, Institute of Physiology, University of Tuebingen, Tuebingen, Germany
| | - Cris Richter
- Department of Neurophysiology, Institute of Physiology, University of Tuebingen, Tuebingen, Germany
| | - Annika Speidel
- Department of Neurophysiology, Institute of Physiology, University of Tuebingen, Tuebingen, Germany
| | - Elizabeta Zirdum
- Department of Neurophysiology, Institute of Physiology, University of Tuebingen, Tuebingen, Germany
| | - Michael Duszenko
- Department of Neurophysiology, Institute of Physiology, University of Tuebingen, Tuebingen, Germany
| | - Olga Garaschuk
- Department of Neurophysiology, Institute of Physiology, University of Tuebingen, Tuebingen, Germany
| | - Katherine Figarella
- Department of Neurophysiology, Institute of Physiology, University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
3
|
Usende IL, Olopade JO, Azeez IA, Andrioli A, Bankole MO, Olopade FE, Nafady AA, Bentivoglio M. Neuroecotoxicology: Effects of environmental heavy metal exposure on the brain of African giant rats and the contribution of vanadium to the neuropathology. IBRO Neurosci Rep 2022; 13:215-234. [PMID: 36590095 PMCID: PMC9795313 DOI: 10.1016/j.ibneur.2022.08.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 08/26/2022] [Indexed: 01/04/2023] Open
Abstract
Increased exploitation of minerals has led to pollution of confined environments as documented in Nigeria Niger Delta. Information on the effects on brain of such exposure is limited. Due to its exploratory activities, the African giant rat (Cricetomys gambianus) (AGR) provides a unique model for neuroecotoxicological research to determine levels of animal and human exposure to different pollutants. This study aims to unravel neuropathological features of AGR sampled from three agro-ecological zones of Nigeria. Fifteen AGR were sampled according to previously determined data on heavy metal exposure: high vanadium, high lead, and low metals. Eighteen AGR were collected from low metal zone and divided into two groups. Control group received vehicle while SMV exposed group received 3 mg/kg sodium metavanadate (SMV) intraperitoneally for 14days. Brain immunohistochemical analyses were conducted, and ultrastructural changes were studied in experimentally exposed group. Results showed significant loss of tyrosin hydroxylase, parvalbumin, orexin-A and melanin concentration hormone containing neuronal populations in brains obtained from high vanadium and high lead zones and in experimentally intoxicated SMV groups. Similarly, significant decrease numbers of dendritic arborations; extracellular matrix density, perineuronal nets; astrocytes and microglia activations are documented in same groups. Ultrastructural studies revealed mass denudation, cilia loss, disintegration of ependymal layer and intense destructions of myelin sheaths in SMV exposed group. These are the first "neuroecotoxicological" findings in distinct neuronal cells. The implications of these findings are highly relevant for human population living in these areas, not only in Nigeria but also in similarly polluted areas elsewhere in the world.
Collapse
Affiliation(s)
- Ifukibot Levi Usende
- Department of Veterinary Anatomy, University of Abuja, Nigeria,Department of Veterinary Anatomy, University of Ibadan, Nigeria,Corresponding author at: Department of Veterinary Anatomy, University of Abuja, Nigeria.
| | | | | | - Anna Andrioli
- Department of Neurological and Movement Sciences, University of Verona, Italy
| | - Molakun O. Bankole
- Department of Neurological and Movement Sciences, University of Verona, Italy
| | | | - Allam A. Nafady
- Department of Veterinary Pathology, Electron Microscope Unit, Assuit University, Egypt
| | - Marina Bentivoglio
- Department of Neurological and Movement Sciences, University of Verona, Italy
| |
Collapse
|
4
|
Olivera GC, Vetter L, Tesoriero C, Del Gallo F, Hedberg G, Basile J, Rottenberg ME. Role of T cells during the cerebral infection with Trypanosoma brucei. PLoS Negl Trop Dis 2021; 15:e0009764. [PMID: 34587172 PMCID: PMC8530334 DOI: 10.1371/journal.pntd.0009764] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 10/21/2021] [Accepted: 08/25/2021] [Indexed: 11/18/2022] Open
Abstract
The infection by Trypanosoma brucei brucei (T.b.b.), a protozoan parasite, is characterized by an early-systemic stage followed by a late stage in which parasites invade the brain parenchyma in a T cell-dependent manner. Here we found that early after infection effector-memory T cells were predominant among brain T cells, whereas, during the encephalitic stage T cells acquired a tissue resident memory phenotype (TRM) and expressed PD1. Both CD4 and CD8 T cells were independently redundant for the penetration of T.b.b. and other leukocytes into the brain parenchyma. The role of lymphoid cells during the T.b.b. infection was studied by comparing T- and B-cell deficient rag1-/- and WT mice. Early after infection, parasites located in circumventricular organs, brain structures with increased vascular permeability, particularly in the median eminence (ME), paced closed to the sleep-wake regulatory arcuate nucleus of the hypothalamus (Arc). Whereas parasite levels in the ME were higher in rag1-/- than in WT mice, leukocytes were instead reduced. Rag1-/- infected mice showed increased levels of meca32 mRNA coding for a blood /hypothalamus endothelial molecule absent in the blood-brain-barrier (BBB). Both immune and metabolic transcripts were elevated in the ME/Arc of WT and rag1-/- mice early after infection, except for ifng mRNA, which levels were only increased in WT mice. Finally, using a non-invasive sleep-wake cycle assessment method we proposed a putative role of lymphocytes in mediating sleep alterations during the infection with T.b.b. Thus, the majority of T cells in the brain during the early stage of T.b.b. infection expressed an effector-memory phenotype while TRM cells developed in the late stage of infection. T cells and parasites invade the ME/Arc altering the metabolic and inflammatory responses during the early stage of infection and modulating sleep disturbances. Trypanosoma brucei (T.b.) causes an early systemic and a late encephalitic infection characterized by sleep alterations. In rodent models, brain invasion by T.b. brucei (T.b.b.) is strictly dependent on T cells. However, an in-depth characterization of T cell functions and phenotypes in the outcome of T.b.b. infection is still lacking. Here we found that during the early stage of infection of mice, most brain T cells differentiated into memory cells, and acquired a tissue-resident memory phenotype during the encephalitic stage. CD4 and CD8 T cells were redundant for the invasion of other T cells and parasites into the brain. Early after infection T.b.b. and leukocytes invade different circumventricular organs (brain areas that lack a blood-brain barrier) including the median eminence (ME) located close to sleep-regulating arcuate nucleus (Arc). T.b.b. infection induced the expression of immune and metabolic molecules in this area. Lymphocytes modulated 1) the levels of invading parasites and leukocytes in the ME; 2) the structure of the blood/ hypothalamus interphase and 3) the expression of IFN-γ in the ME/Arc early after infection. Lymphocytes may also be involved in the regulation of sleep alterations observed in African trypanosomiasis.
Collapse
Affiliation(s)
- Gabriela C. Olivera
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Leonie Vetter
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Chiara Tesoriero
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Federico Del Gallo
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Gustav Hedberg
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Juan Basile
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Martin E. Rottenberg
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
5
|
Rijo-Ferreira F, Bjorness TE, Cox KH, Sonneborn A, Greene RW, Takahashi JS. Sleeping Sickness Disrupts the Sleep-Regulating Adenosine System. J Neurosci 2020; 40:9306-9316. [PMID: 33097636 PMCID: PMC7687053 DOI: 10.1523/jneurosci.1046-20.2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 09/28/2020] [Accepted: 10/11/2020] [Indexed: 12/16/2022] Open
Abstract
Patients with sleeping sickness, caused by the parasite Trypanosoma brucei, have disruptions in both sleep timing and sleep architecture. However, the underlying cause of these sleep disturbances is not well understood. Here, we assessed the sleep architecture of male mice infected with T. brucei and found that infected mice had drastically altered sleep patterns. Interestingly, T. brucei-infected mice also had a reduced homeostatic sleep response to sleep deprivation, a response modulated by the adenosine system. We found that infected mice had a reduced electrophysiological response to an adenosine receptor antagonist and increased adenosine receptor gene expression. Although the mechanism by which T. brucei infection causes these changes remains to be determined, our findings suggest that the symptoms of sleeping sickness may be because of alterations in homeostatic adenosine signaling.SIGNIFICANCE STATEMENT Sleeping sickness is a fatal disease that disrupts the circadian clock, causes disordered temperature regulation, and induces sleep disturbance. To examine the neurologic effects of infection in the absence of other symptoms, in this study, we used a mouse model of sleeping sickness in which the acute infection was treated but brain infection remained. Using this model, we evaluated the effects of the sleeping sickness parasite, Trypanosoma brucei, on sleep patterns in mice, under both normal and sleep-deprived conditions. Our findings suggest that signaling of adenosine, a neuromodulator involved in mediating homeostatic sleep drive, may be reduced in infected mice.
Collapse
Affiliation(s)
- Filipa Rijo-Ferreira
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9111
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9111
| | - Theresa E Bjorness
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9111
- Research Service, VA North Texas Health Care System, Dallas, Texas 75216-7167
| | - Kimberly H Cox
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9111
| | - Alex Sonneborn
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9111
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9111
| | - Robert W Greene
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9111
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9111
| | - Joseph S Takahashi
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9111
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9111
| |
Collapse
|
6
|
Rijo-Ferreira F, Takahashi JS. Sleeping Sickness: A Tale of Two Clocks. Front Cell Infect Microbiol 2020; 10:525097. [PMID: 33134186 PMCID: PMC7562814 DOI: 10.3389/fcimb.2020.525097] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 08/31/2020] [Indexed: 12/17/2022] Open
Abstract
Sleeping sickness is caused by a eukaryotic unicellular parasite known to infect wild animals, cattle, and humans. It causes a fatal disease that disrupts many rhythmic physiological processes, including daily rhythms of hormonal secretion, temperature regulation, and sleep, all of which are under circadian (24-h) control. In this review, we summarize research on sleeping sickness parasite biology and the impact it has on host health. We also consider the possible evolutionary advantages of sleep and circadian deregulation for the parasite.
Collapse
Affiliation(s)
- Filipa Rijo-Ferreira
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, United States.,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Joseph S Takahashi
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, United States.,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
7
|
The ameliorative effects of a phenolic derivative of Moringa oleifera leave against vanadium-induced neurotoxicity in mice. IBRO Rep 2020; 9:164-182. [PMID: 32803016 PMCID: PMC7417907 DOI: 10.1016/j.ibror.2020.07.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 07/03/2020] [Indexed: 12/29/2022] Open
Abstract
Vanadium, a transition series metal released during some industrial activities, induces oxidative stress and lipid peroxidation. Ameliorative effect of a pure compound from the methanolic extract of Moringa oleifera leaves, code-named MIMO2, in 14-day old mice administered with vanadium (as sodium metavanadate 3 mg/kg) for 2 weeks was assessed. Results from body weight monitoring, muscular strength, and open field showed slight reduction in body weight and locomotion deficit in vanadium-exposed mice, ameliorated with MIMO2 co-administration. Degeneration of the Purkinje cell layer and neuronal death in the hippocampal CA1 region were observed in vanadium-exposed mice and both appeared significantly reduced with MIMO2 co-administration. Demyelination involving the midline of the corpus callosum, somatosensory and retrosplenial cortices was also reduced with MIMO2. Microglia activation and astrogliosis observed through immunohistochemistry were also alleviated. Immunohistochemistry for myelin, axons and oligodendrocyte lineage cells were also carried out and showed that in vanadium-treated mice brains, oligodendrocyte progenitor cells increased NG2 immunolabelling with hypertrophy and bushy, ramified appearance of their processes. MIMO2 displayed ameliorative and antioxidative effects in vanadium-induced neurotoxicity in experimental murine species. This is likely the first time MIMO2 is being used in vivo in an animal model.
Collapse
|
8
|
Bentivoglio M, Kristensson K, Rottenberg ME. Circumventricular Organs and Parasite Neurotropism: Neglected Gates to the Brain? Front Immunol 2018; 9:2877. [PMID: 30619260 PMCID: PMC6302769 DOI: 10.3389/fimmu.2018.02877] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 11/22/2018] [Indexed: 12/20/2022] Open
Abstract
Circumventricular organs (CVOs), neural structures located around the third and fourth ventricles, harbor, similarly to the choroid plexus, vessels devoid of a blood-brain barrier (BBB). This enables them to sense immune-stimulatory molecules in the blood circulation, but may also increase chances of exposure to microbes. In spite of this, attacks to CVOs by microbes are rarely described. It is here highlighted that CVOs and choroid plexus can be infected by pathogens circulating in the bloodstream, providing a route for brain penetration, as shown by infections with the parasites Trypanosoma brucei. Immune responses elicited by pathogens or systemic infections in the choroid plexus and CVOs are briefly outlined. From the choroid plexus trypanosomes can seed into the ventricles and initiate accelerated infiltration of T cells and parasites in periventricular areas. The highly motile trypanosomes may also enter the brain parenchyma from the median eminence, a CVO located at the base of the third ventricle, by crossing the border into the BBB-protected hypothalamic arcuate nuclei. A gate may, thus, be provided for trypanosomes to move into brain areas connected to networks of regulation of circadian rhythms and sleep-wakefulness, to which other CVOs are also connected. Functional imbalances in these networks characterize human African trypanosomiasis, also called sleeping sickness. They are distinct from the sickness response to bacterial infections, but can occur in common neuropsychiatric diseases. Altogether the findings lead to the question: does the neglect in reporting microbe attacks to CVOs reflect lack of awareness in investigations or of gate-opening capability by microbes?
Collapse
Affiliation(s)
- Marina Bentivoglio
- Department of Neuroscience Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | | | - Martin E. Rottenberg
- Department Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
9
|
Tesoriero C, Del Gallo F, Bentivoglio M. Sleep and brain infections. Brain Res Bull 2018; 145:59-74. [PMID: 30016726 DOI: 10.1016/j.brainresbull.2018.07.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 07/10/2018] [Accepted: 07/12/2018] [Indexed: 12/11/2022]
Abstract
Sleep is frequently altered in systemic infections as a component of sickness behavior in response to inflammation. Sleepiness in sickness behavior has been extensively investigated. Much less attention has instead been devoted to sleep and wake alterations in brain infections. Most of these, as other neuroinfections, are prevalent in sub-Saharan Africa. The present overview highlights the importance of this topic from both the clinical and pathogenetic points of view. Vigilance states and their regulation are first summarized, emphasizing that key nodes in this distributed brain system can be targeted by neuroinflammatory signaling. Sleep-wake changes in the parasitic disease human African trypanosomiasis (HAT) and its animal models are then reviewed and discussed. Experimental data have revealed that the suprachiasmatic nucleus, the master circadian pacemaker, and peptidergic cell populations of the lateral hypothalamus (the wake-promoting orexin neurons and the sleep-promoting melanin-concentrating hormone neurons) are targeted by African trypanosome infection. It is then discussed how prominent and disturbing are sleep changes in HIV/AIDS, also when the infection is cured with antiretroviral therapy. This recalls attention on the bidirectional interactions between sleep and immune system, including the specialized brain immune response of which microglial cells are protagonists. Sleep changes in an ancient viral disease, rabies, and in the emerging infection due to Zika virus which causes a congenital syndrome, are also dealt with. Altogether the findings indicate that sleep-wake regulation is targeted by brain infections caused by different pathogens and, although the relevant pathogenetic mechanisms largely remain to be clarified, these alterations differ from hypersomnia occurring in sickness behavior. Thus, brain infections point to the vulnerability of the neural network of sleep-wake regulation as a highly relevant clinical and basic science challenge.
Collapse
Affiliation(s)
- Chiara Tesoriero
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy
| | - Federico Del Gallo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy
| | - Marina Bentivoglio
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy.
| |
Collapse
|
10
|
Hopwood TW, Hall S, Begley N, Forman R, Brown S, Vonslow R, Saer B, Little MC, Murphy EA, Hurst RJ, Ray DW, MacDonald AS, Brass A, Bechtold DA, Gibbs JE, Loudon AS, Else KJ. The circadian regulator BMAL1 programmes responses to parasitic worm infection via a dendritic cell clock. Sci Rep 2018; 8:3782. [PMID: 29491349 PMCID: PMC5830501 DOI: 10.1038/s41598-018-22021-5] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 02/14/2018] [Indexed: 12/13/2022] Open
Abstract
Resistance to the intestinal parasitic helminth Trichuris muris requires T-helper 2 (TH2) cellular and associated IgG1 responses, with expulsion typically taking up to 4 weeks in mice. Here, we show that the time-of-day of the initial infection affects efficiency of worm expulsion, with strong TH2 bias and early expulsion in morning-infected mice. Conversely, mice infected at the start of the night show delayed resistance to infection, and this is associated with feeding-driven metabolic cues, such that feeding restriction to the day-time in normally nocturnal-feeding mice disrupts parasitic expulsion kinetics. We deleted the circadian regulator BMAL1 in antigen-presenting dendritic cells (DCs) in vivo and found a loss of time-of-day dependency of helminth expulsion. RNAseq analyses revealed that IL-12 responses to worm antigen by circadian-synchronised DCs were dependent on BMAL1. Therefore, we find that circadian machinery in DCs contributes to the TH1/TH2 balance, and that environmental, or genetic perturbation of the DC clock results in altered parasite expulsion kinetics.
Collapse
Affiliation(s)
- Thomas W Hopwood
- Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
- Manchester Academic Health Sciences Centre, Manchester, United Kingdom
| | - Sarah Hall
- Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
- Manchester Academic Health Sciences Centre, Manchester, United Kingdom
| | - Nicola Begley
- Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
- Manchester Academic Health Sciences Centre, Manchester, United Kingdom
| | - Ruth Forman
- Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
- Manchester Academic Health Sciences Centre, Manchester, United Kingdom
| | - Sheila Brown
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, M13 9NT, UK
| | - Ryan Vonslow
- Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
- Manchester Academic Health Sciences Centre, Manchester, United Kingdom
| | - Ben Saer
- Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
- Manchester Academic Health Sciences Centre, Manchester, United Kingdom
| | - Matthew C Little
- Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
- Manchester Academic Health Sciences Centre, Manchester, United Kingdom
| | - Emma A Murphy
- Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
- Manchester Academic Health Sciences Centre, Manchester, United Kingdom
| | - Rebecca J Hurst
- Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
- Manchester Academic Health Sciences Centre, Manchester, United Kingdom
| | - David W Ray
- Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
- Manchester Academic Health Sciences Centre, Manchester, United Kingdom
| | - Andrew S MacDonald
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, M13 9NT, UK
| | - Andy Brass
- Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
- Manchester Academic Health Sciences Centre, Manchester, United Kingdom
| | - David A Bechtold
- Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
- Manchester Academic Health Sciences Centre, Manchester, United Kingdom
| | - Julie E Gibbs
- Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, M13 9PT, UK.
- Manchester Academic Health Sciences Centre, Manchester, United Kingdom.
| | - Andrew S Loudon
- Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, M13 9PT, UK.
- Manchester Academic Health Sciences Centre, Manchester, United Kingdom.
| | - Kathryn J Else
- Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, M13 9PT, UK.
- Manchester Academic Health Sciences Centre, Manchester, United Kingdom.
| |
Collapse
|
11
|
Laperchia C, Xu YZ, Mumba Ngoyi D, Cotrufo T, Bentivoglio M. Neural Damage in Experimental Trypanosoma brucei gambiense Infection: Hypothalamic Peptidergic Sleep and Wake-Regulatory Neurons. Front Neuroanat 2018. [PMID: 29535612 PMCID: PMC5835115 DOI: 10.3389/fnana.2018.00013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Neuron populations of the lateral hypothalamus which synthesize the orexin (OX)/hypocretin or melanin-concentrating hormone (MCH) peptides play crucial, reciprocal roles in regulating wake stability and sleep. The disease human African trypanosomiasis (HAT), also called sleeping sickness, caused by extracellular Trypanosoma brucei (T. b.) parasites, leads to characteristic sleep-wake cycle disruption and narcoleptic-like alterations of the sleep structure. Previous studies have revealed damage of OX and MCH neurons during systemic infection of laboratory rodents with the non-human pathogenic T. b. brucei subspecies. No information is available, however, on these peptidergic neurons after systemic infection with T. b. gambiense, the etiological agent of 97% of HAT cases. The present study was aimed at the investigation of immunohistochemically characterized OX and MCH neurons after T. b. gambiense or T. b. brucei infection of a susceptible rodent, the multimammate mouse, Mastomysnatalensis. Cell counts and evaluation of OX fiber density were performed at 4 and 8 weeks post-infection, when parasites had entered the brain parenchyma from the periphery. A significant decrease of OX neurons (about 44% reduction) and MCH neurons (about 54% reduction) was found in the lateral hypothalamus and perifornical area at 8 weeks in T. b. gambiense-infected M. natalensis. A moderate decrease (21% and 24% reduction, respectively), which did not reach statistical significance, was found after T. b. brucei infection. In two key targets of diencephalic orexinergic innervation, the peri-suprachiasmatic nucleus (SCN) region and the thalamic paraventricular nucleus (PVT), densitometric analyses showed a significant progressive decrease in the density of orexinergic fibers in both infection paradigms, and especially during T. b. gambiense infection. Altogether the findings provide novel information showing that OX and MCH neurons are highly vulnerable to chronic neuroinflammatory signaling caused by the infection of human-pathogenic African trypanosomes.
Collapse
Affiliation(s)
- Claudia Laperchia
- Department of Neuroscience Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Yuan-Zhong Xu
- Department of Neuroscience Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Dieudonné Mumba Ngoyi
- Institut National de Recherche Biomédicale (INRB), Kinshasa, Democratic Republic of Congo
| | - Tiziana Cotrufo
- Department of Neuroscience Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Marina Bentivoglio
- Department of Neuroscience Biomedicine and Movement Sciences, University of Verona, Verona, Italy.,National Institute of Neuroscience (INN), Verona Unit, Verona, Italy
| |
Collapse
|
12
|
Laperchia C, Tesoriero C, Seke-Etet PF, La Verde V, Colavito V, Grassi-Zucconi G, Rodgers J, Montague P, Kennedy PGE, Bentivoglio M. Expression of interferon-inducible chemokines and sleep/wake changes during early encephalitis in experimental African trypanosomiasis. PLoS Negl Trop Dis 2017; 11:e0005854. [PMID: 28821016 PMCID: PMC5576758 DOI: 10.1371/journal.pntd.0005854] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 08/30/2017] [Accepted: 08/04/2017] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Human African trypanosomiasis or sleeping sickness, caused by the parasite Trypanosoma brucei, leads to neuroinflammation and characteristic sleep/wake alterations. The relationship between the onset of these alterations and the development of neuroinflammation is of high translational relevance, but remains unclear. This study investigates the expression of interferon (IFN)-γ and IFN-inducible chemokine genes in the brain, and the levels of CXCL10 in the serum and cerebrospinal fluid prior to and during the encephalitic stage of trypanosome infection, and correlates these with sleep/wake changes in a rat model of the disease. METHODOLOGY/PRINCIPAL FINDINGS The expression of genes encoding IFN-γ, CXCL9, CXCL10, and CXCL11 was assessed in the brain of rats infected with Trypanosoma brucei brucei and matched controls using semi-quantitative end-point RT-PCR. Levels of CXCL10 in the serum and cerebrospinal fluid were determined using ELISA. Sleep/wake states were monitored by telemetric recording. Using immunohistochemistry, parasites were found in the brain parenchyma at 14 days post-infection (dpi), but not at 6 dpi. Ifn-γ, Cxcl9, Cxcl10 and Cxcl11 mRNA levels showed moderate upregulation by 14 dpi followed by further increase between 14 and 21 dpi. CXCL10 concentration in the cerebrospinal fluid increased between 14 and 21 dpi, preceded by a rise in the serum CXCL10 level between 6 and 14 dpi. Sleep/wake pattern fragmentation was evident at 14 dpi, especially in the phase of wake predominance, with intrusion of sleep episodes into wakefulness. CONCLUSIONS/SIGNIFICANCE The results show a modest increase in Cxcl9 and Cxcl11 transcripts in the brain and the emergence of sleep/wake cycle fragmentation in the initial encephalitic stage, followed by increases in Ifn-γ and IFN-dependent chemokine transcripts in the brain and of CXCL10 in the cerebrospinal fluid. The latter parameter and sleep/wake alterations could provide combined humoral and functional biomarkers of the early encephalitic stage in African trypanosomiasis.
Collapse
Affiliation(s)
- Claudia Laperchia
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Chiara Tesoriero
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Paul F. Seke-Etet
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Valentina La Verde
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Valeria Colavito
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Gigliola Grassi-Zucconi
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Jean Rodgers
- Institute of Biodiversity, Animal Health & Comparative Medicine, University of Glasgow, Glasgow, United Kingdom
| | - Paul Montague
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Peter G. E. Kennedy
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, United Kingdom
- * E-mail:
| | - Marina Bentivoglio
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
- National Institute of Neuroscience (INN), Verona Unit, Verona, Italy
| |
Collapse
|
13
|
The excitatory/inhibitory input to orexin/hypocretin neuron soma undergoes day/night reorganization. Brain Struct Funct 2017; 222:3847-3859. [PMID: 28669028 DOI: 10.1007/s00429-017-1466-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 06/20/2017] [Indexed: 02/07/2023]
Abstract
Orexin (OX)/hypocretin-containing neurons are main regulators of wakefulness stability, arousal, and energy homeostasis. Their activity varies in relation to the animal's behavioral state. We here tested whether such variation is subserved by synaptic plasticity phenomena in basal conditions. Mice were sacrificed during day or night, at times when sleep or wake, respectively, predominates, as assessed by electroencephalography in matched mice. Triple immunofluorescence was used to visualize OX-A perikarya and varicosities containing the vesicular glutamate transporter (VGluT)2 or the vesicular GABA transporter (VGAT) combined with synaptophysin (Syn) as a presynaptic marker. Appositions on OX-A+ somata were quantitatively analyzed in pairs of sections in epifluorescence and confocal microscopy. The combined total number of glutamatergic (Syn+/VGluT2+) and GABAergic (Syn+/VGAT+) varicosities apposed to OX-A somata was similar during day and night. However, glutamatergic varicosities were significantly more numerous at night, whereas GABAergic varicosities prevailed in the day. Triple immunofluorescence in confocal microscopy was employed to visualize synapse scaffold proteins as postsynaptic markers and confirmed the nighttime prevalence of VGluT2+ together with postsynaptic density protein 95+ excitatory contacts, and daytime prevalence of VGAT+ together with gephyrin+ inhibitory contacts, while also showing that they formed synapses on OX-A+ cell bodies. The findings reveal a daily reorganization of axosomatic synapses in orexinergic neurons, with a switch from a prevalence of excitatory innervation at a time corresponding to wakefulness to a prevalence of inhibitory innervations in the antiphase, at a time corresponding to sleep. This reorganization could represent a key mechanism of plasticity of the orexinergic network in basal conditions.
Collapse
|
14
|
Diurnal fluctuation in the number of hypocretin/orexin and histamine producing: Implication for understanding and treating neuronal loss. PLoS One 2017; 12:e0178573. [PMID: 28570646 PMCID: PMC5453544 DOI: 10.1371/journal.pone.0178573] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Accepted: 05/15/2017] [Indexed: 11/24/2022] Open
Abstract
The loss of specific neuronal phenotypes, as determined by immunohistochemistry, has become a powerful tool for identifying the nature and cause of neurological diseases. Here we show that the number of neurons identified and quantified using this method misses a substantial percentage of extant neurons in a phenotype specific manner. In mice, 24% more hypocretin/orexin (Hcrt) neurons are seen in the night compared to the day, and an additional 17% are seen after inhibiting microtubule polymerization with colchicine. We see no such difference between the number of MCH (melanin concentrating hormone) neurons in dark, light or colchicine conditions, despite MCH and Hcrt both being hypothalamic peptide transmitters. Although the size of Hcrt neurons did not differ between light and dark, the size of MCH neurons was increased by 15% in the light phase. The number of neurons containing histidine decarboxylase (HDC), the histamine synthesizing enzyme, was 34% greater in the dark than in the light, but, like Hcrt, cell size did not differ. We did not find a significant difference in the number or the size of neurons expressing choline acetyltransferase (ChAT), the acetylcholine synthesizing enzyme, in the horizontal diagonal band (HBD) during the dark and light conditions. As expected, colchicine treatment did not increase the number of these neurons. Understanding the function and dynamics of transmitter production within “non-visible” phenotypically defined cells has fundamental implications for our understanding of brain plasticity.
Collapse
|
15
|
Laperchia C, Palomba M, Seke Etet PF, Rodgers J, Bradley B, Montague P, Grassi-Zucconi G, Kennedy PGE, Bentivoglio M. Trypanosoma brucei Invasion and T-Cell Infiltration of the Brain Parenchyma in Experimental Sleeping Sickness: Timing and Correlation with Functional Changes. PLoS Negl Trop Dis 2016; 10:e0005242. [PMID: 28002454 PMCID: PMC5217973 DOI: 10.1371/journal.pntd.0005242] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 01/06/2017] [Accepted: 12/07/2016] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The timing of Trypanosoma brucei entry into the brain parenchyma to initiate the second, meningoencephalitic stage of human African trypanosomiasis or sleeping sickness is currently debated and even parasite invasion of the neuropil has been recently questioned. Furthermore, the relationship between neurological features and disease stage are unclear, despite the important diagnostic and therapeutic implications. METHODOLOGY Using a rat model of chronic Trypanosoma brucei brucei infection we determined the timing of parasite and T-cell neuropil infiltration and its correlation with functional changes. Parasite DNA was detected using trypanosome-specific PCR. Body weight and sleep structure alterations represented by sleep-onset rapid eye movement (SOREM) periods, reported in human and experimental African trypanosomiasis, were monitored. The presence of parasites, as well as CD4+ and CD8+ T-cells in the neuropil was assessed over time in the brain of the same animals by immunocytochemistry and quantitative analyses. PRINCIPAL FINDINGS Trypanosome DNA was present in the brain at day 6 post-infection and increased more than 15-fold by day 21. Parasites and T-cells were observed in the parenchyma from day 9 onwards. Parasites traversing blood vessel walls were observed in the hypothalamus and other brain regions. Body weight gain was reduced from day 7 onwards. SOREM episodes started in most cases early after infection, with an increase in number and duration after parasite neuroinvasion. CONCLUSION These findings demonstrate invasion of the neuropil over time, after an initial interval, by parasites and lymphocytes crossing the blood-brain barrier, and show that neurological features can precede this event. The data thus challenge the current clinical and cerebrospinal fluid criteria of disease staging.
Collapse
Affiliation(s)
- Claudia Laperchia
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Maria Palomba
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Paul F. Seke Etet
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Jean Rodgers
- Institute of Biodiversity Animal Health and Comparative Medicine, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Barbara Bradley
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Paul Montague
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Gigliola Grassi-Zucconi
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Peter G. E. Kennedy
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Marina Bentivoglio
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
- National Institute of Neuroscience (INN), Verona Unit, Verona, Italy
| |
Collapse
|
16
|
Azeez IA, Olopade F, Laperchia C, Andrioli A, Scambi I, Onwuka SK, Bentivoglio M, Olopade JO. Regional Myelin and Axon Damage and Neuroinflammation in the Adult Mouse Brain After Long-Term Postnatal Vanadium Exposure. J Neuropathol Exp Neurol 2016; 75:843-54. [PMID: 27390101 DOI: 10.1093/jnen/nlw058] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Indexed: 01/08/2023] Open
Abstract
Environmental exposure to vanadium occurs in areas of persistent burning of fossil fuels; this metal is known to induce oxidative stress and oligodendrocyte damage. Here, we determined whether vanadium exposure (3 mg/kg) in mice during the first 3 postnatal months leads to a sustained neuroinflammatory response. Body weight monitoring, and muscle strength and open field tests showed reduction of body weight gain and locomotor impairment in vanadium-exposed mice. Myelin histochemistry and immunohistochemistry for astrocytes, microglia, and nonphosphorylated neurofilaments revealed striking regional heterogeneity. Myelin damage involved the midline corpus callosum and fibers in cortical gray matter, hippocampus, and diencephalon that were associated with axonal damage. Astrocyte and microglial activation was identified in the same regions and in the internal capsule; however, no overt myelin and axon damage was observed in the latter. Double immunofluorescence revealed induction of high tumor necrosis factor (TNF) immunoreactivity in reactive astrocytes. Western blotting analysis showed significant induction of TNF and interleukin-1β expression. Together these findings show that chronic postnatal vanadium exposure leads to functional deficit and region-dependent myelin damage that does not spare axons. This injury is associated with glial cell activation and proinflammatory cytokine induction, which may reflect both neurotoxic and neuroprotective responses.
Collapse
Affiliation(s)
- Idris A Azeez
- From the Department of Veterinary Anatomy, University of Ibadan, Nigeria (IAA, SKO, JOO); Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Italy (IAA, CL, AA, IS, MB); Department of Anatomy, University of Ibadan, Nigeria (FO)
| | - Funmilayo Olopade
- From the Department of Veterinary Anatomy, University of Ibadan, Nigeria (IAA, SKO, JOO); Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Italy (IAA, CL, AA, IS, MB); Department of Anatomy, University of Ibadan, Nigeria (FO)
| | - Claudia Laperchia
- From the Department of Veterinary Anatomy, University of Ibadan, Nigeria (IAA, SKO, JOO); Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Italy (IAA, CL, AA, IS, MB); Department of Anatomy, University of Ibadan, Nigeria (FO)
| | - Anna Andrioli
- From the Department of Veterinary Anatomy, University of Ibadan, Nigeria (IAA, SKO, JOO); Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Italy (IAA, CL, AA, IS, MB); Department of Anatomy, University of Ibadan, Nigeria (FO)
| | - Ilaria Scambi
- From the Department of Veterinary Anatomy, University of Ibadan, Nigeria (IAA, SKO, JOO); Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Italy (IAA, CL, AA, IS, MB); Department of Anatomy, University of Ibadan, Nigeria (FO)
| | - Silas K Onwuka
- From the Department of Veterinary Anatomy, University of Ibadan, Nigeria (IAA, SKO, JOO); Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Italy (IAA, CL, AA, IS, MB); Department of Anatomy, University of Ibadan, Nigeria (FO)
| | - Marina Bentivoglio
- From the Department of Veterinary Anatomy, University of Ibadan, Nigeria (IAA, SKO, JOO); Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Italy (IAA, CL, AA, IS, MB); Department of Anatomy, University of Ibadan, Nigeria (FO)
| | - James O Olopade
- From the Department of Veterinary Anatomy, University of Ibadan, Nigeria (IAA, SKO, JOO); Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Italy (IAA, CL, AA, IS, MB); Department of Anatomy, University of Ibadan, Nigeria (FO).
| |
Collapse
|