Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.579
Filter
1.
Biomark Med ; 18(9): 431-439, 2024.
Article in English | MEDLINE | ID: mdl-39007837

ABSTRACT

Leptomeningeal metastasis (LM) is a devastating complication of malignancy. Diagnosis relies on both contrast enhancement on imaging and malignant cells in cerebral spinal fluid cytology. Though early detection and prompt intervention improves survival, the detection of LM is limited by false negatives. A rare brainstem imaging finding uncovered specifically in EGFR mutation-positive lung cancer patients may represent an early sign of LM. This sign demonstrates high signal on T2 fluid-attenuated inversion recovery and diffusion-weighted imaging sequences, but paradoxically lacks correlative contrast enhancement. Here we report a case of a 72-year-old female EGFR-positive lung cancer patient who developed this lesion following treatment with two first-generation EGFR tyrosine kinase inhibitors then showed subsequent response to osimertinib, an irreversible third-generation EGFR tyrosine kinase inhibitor.


A non-enhancing, T2 FLAIR hyperintense, diffusion-restricting brainstem lesion in an EGFR-positive lung cancer patient may represent an early indicator of leptomeningeal metastases.


Subject(s)
Carcinoma, Non-Small-Cell Lung , ErbB Receptors , Lung Neoplasms , Protein Kinase Inhibitors , Humans , Female , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Carcinoma, Non-Small-Cell Lung/metabolism , Aged , Protein Kinase Inhibitors/therapeutic use , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/diagnostic imaging , Brain Stem/pathology , Brain Stem/diagnostic imaging , Brain Stem/metabolism , Aniline Compounds/therapeutic use , Acrylamides/therapeutic use , Diffusion Magnetic Resonance Imaging , Indoles , Pyrimidines
2.
Sci Rep ; 14(1): 14734, 2024 06 26.
Article in English | MEDLINE | ID: mdl-38926520

ABSTRACT

Based on the auditory periphery and the small head size, Etruscan shrews (Suncus etruscus) approximate ancestral mammalian conditions. The auditory brainstem in this insectivore has not been investigated. Using labelling techniques, we assessed the structures of their superior olivary complex (SOC) and the nuclei of the lateral lemniscus (NLL). There, we identified the position of the major nuclei, their input pattern, transmitter content, expression of calcium binding proteins (CaBPs) and two voltage-gated ion channels. The most prominent SOC structures were the medial nucleus of the trapezoid body (MNTB), the lateral nucleus of the trapezoid body (LNTB), the lateral superior olive (LSO) and the superior paraolivary nucleus (SPN). In the NLL, the ventral (VNLL), a specific ventrolateral VNLL (VNLLvl) cell population, the intermediate (INLL) and dorsal (DNLL) nucleus, as well as the inferior colliculus's central aspect were discerned. INLL and VNLL were clearly separated by the differential distribution of various marker proteins. Most labelled proteins showed expression patterns comparable to rodents. However, SPN neurons were glycinergic and not GABAergic and the overall CaBPs expression was low. Next to the characterisation of the Etruscan shrew's auditory brainstem, our work identifies conserved nuclei and indicates variable structures in a species that approximates ancestral conditions.


Subject(s)
Shrews , Superior Olivary Complex , Animals , Shrews/anatomy & histology , Superior Olivary Complex/anatomy & histology , Superior Olivary Complex/metabolism , Auditory Pathways/anatomy & histology , Neurons/metabolism , Inferior Colliculi/anatomy & histology , Inferior Colliculi/metabolism , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/genetics , Brain Stem/anatomy & histology , Brain Stem/metabolism , Male , Olivary Nucleus/anatomy & histology , Olivary Nucleus/metabolism
3.
Nat Commun ; 15(1): 5133, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38879548

ABSTRACT

Lewy body (LB) diseases, characterized by the aggregation of misfolded α-synuclein proteins, exhibit notable clinical heterogeneity. This may be due to variations in accumulation patterns of LB neuropathology. Here we apply a data-driven disease progression model to regional neuropathological LB density scores from 814 brain donors with Lewy pathology. We describe three inferred trajectories of LB pathology that are characterized by differing clinicopathological presentation and longitudinal antemortem clinical progression. Most donors (81.9%) show earliest pathology in the olfactory bulb, followed by accumulation in either limbic (60.8%) or brainstem (21.1%) regions. The remaining donors (18.1%) initially exhibit abnormalities in brainstem regions. Early limbic pathology is associated with Alzheimer's disease-associated characteristics while early brainstem pathology is associated with progressive motor impairment and substantial LB pathology outside of the brain. Our data provides evidence for heterogeneity in the temporal spread of LB pathology, possibly explaining some of the clinical disparities observed in Lewy body disease.


Subject(s)
Disease Progression , Lewy Bodies , Lewy Body Disease , alpha-Synuclein , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , alpha-Synuclein/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , Brain/pathology , Brain/metabolism , Brain Stem/pathology , Brain Stem/metabolism , Lewy Bodies/pathology , Lewy Bodies/metabolism , Lewy Body Disease/pathology , Lewy Body Disease/metabolism , Olfactory Bulb/pathology , Olfactory Bulb/metabolism
5.
Int J Mol Sci ; 25(11)2024 May 21.
Article in English | MEDLINE | ID: mdl-38891784

ABSTRACT

The central nervous system of Pacific salmon retains signs of embryonic structure throughout life and a large number of neuroepithelial neural stem cells (NSCs) in the proliferative areas of the brain, in particular. However, the adult nervous system and neurogenesis studies on rainbow trout, Oncorhynchus mykiss, are limited. Here, we studied the localization of glutamine synthetase (GS), vimentin (Vim), and nestin (Nes), as well as the neurons formed in the postembryonic period, labeled with doublecortin (DC), under conditions of homeostatic growth in adult cerebellum and brainstem of Oncorhynchus mykiss using immunohistochemical methods and Western Immunoblotting. We observed that the distribution of vimentin (Vim), nestin (Nes), and glutamine synthetase (GS), which are found in the aNSPCs of both embryonic types (neuroepithelial cells) and in the adult type (radial glia) in the cerebellum and the brainstem of trout, has certain features. Populations of the adult neural stem/progenitor cells (aNSPCs) expressing GS, Vim, and Nes have different morphologies, localizations, and patterns of cluster formation in the trout cerebellum and brainstem, which indicates the morphological and, obviously, functional heterogeneity of these cells. Immunolabeling of PCNA revealed areas in the cerebellum and brainstem of rainbow trout containing proliferating cells which coincide with areas expressing Vim, Nes, and GS. Double immunolabeling revealed the PCNA/GS PCNA/Vim coexpression patterns in the neuroepithelial-type cells in the PVZ of the brainstem. PCNA/GS coexpression in the RG was detected in the submarginal zone of the brainstem. The results of immunohistochemical study of the DC distribution in the cerebellum and brainstem of trout have showed a high level of expression of this marker in various cell populations. This may indicate: (i) high production of the adult-born neurons in the cerebellum and brainstem of adult trout, (ii) high plasticity of neurons in the cerebellum and brainstem of trout. We assume that the source of new cells in the trout brain, along with PVZ and SMZ, containing proliferating cells, may be local neurogenic niches containing the PCNA-positive and silent (PCNA-negative), but expressing NSC markers, cells. The identification of cells expressing DC, Vim, and Nes in the IX-X cranial nerve nuclei of trout was carried out.


Subject(s)
Brain Stem , Cerebellum , Neural Stem Cells , Neurogenesis , Neuronal Plasticity , Oncorhynchus mykiss , Animals , Oncorhynchus mykiss/metabolism , Oncorhynchus mykiss/growth & development , Cerebellum/metabolism , Cerebellum/cytology , Cerebellum/growth & development , Neurogenesis/physiology , Neuronal Plasticity/physiology , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Brain Stem/metabolism , Brain Stem/cytology , Vimentin/metabolism , Neurons/metabolism , Neurons/cytology , Proliferating Cell Nuclear Antigen/metabolism , Glutamate-Ammonia Ligase/metabolism
6.
Elife ; 132024 May 30.
Article in English | MEDLINE | ID: mdl-38814174

ABSTRACT

Neurexins play diverse functions as presynaptic organizers in various glutamatergic and GABAergic synapses. However, it remains unknown whether and how neurexins are involved in shaping functional properties of the glycinergic synapses, which mediate prominent inhibition in the brainstem and spinal cord. To address these issues, we examined the role of neurexins in a model glycinergic synapse between the principal neuron in the medial nucleus of the trapezoid body (MNTB) and the principal neuron in the lateral superior olive (LSO) in the auditory brainstem. Combining RNAscope with stereotactic injection of AAV-Cre in the MNTB of neurexin1/2/3 conditional triple knockout mice, we showed that MNTB neurons highly express all isoforms of neurexins although their expression levels vary remarkably. Selective ablation of all neurexins in MNTB neurons not only reduced the amplitude but also altered the kinetics of the glycinergic synaptic transmission at LSO neurons. The synaptic dysfunctions primarily resulted from an impaired Ca2+ sensitivity of release and a loosened coupling between voltage-gated Ca2+ channels and synaptic vesicles. Together, our current findings demonstrate that neurexins are essential in controlling the strength and temporal precision of the glycinergic synapse, which therefore corroborates the role of neurexins as key presynaptic organizers in all major types of fast chemical synapses.


Subject(s)
Glycine , Mice, Knockout , Trapezoid Body , Animals , Glycine/metabolism , Mice , Trapezoid Body/metabolism , Trapezoid Body/physiology , Synaptic Transmission/physiology , Neural Cell Adhesion Molecules/metabolism , Neural Cell Adhesion Molecules/genetics , Superior Olivary Complex/physiology , Superior Olivary Complex/metabolism , Brain Stem/physiology , Brain Stem/metabolism , Synapses/metabolism , Synapses/physiology , Neurons/metabolism , Neurons/physiology , Cell Adhesion Molecules, Neuronal/metabolism , Cell Adhesion Molecules, Neuronal/genetics , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Neurexins , Calcium-Binding Proteins
7.
J Neurophysiol ; 132(1): 108-129, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38748514

ABSTRACT

µ-Opioid receptors (MORs) are responsible for mediating both the analgesic and respiratory effects of opioid drugs. By binding to MORs in brainstem regions involved in controlling breathing, opioids produce respiratory depressive effects characterized by slow and shallow breathing, with potential cardiorespiratory arrest and death during overdose. To better understand the mechanisms underlying opioid-induced respiratory depression, thorough knowledge of the regions and cellular subpopulations that may be vulnerable to modulation by opioid drugs is needed. Using in situ hybridization, we determined the distribution and coexpression of Oprm1 (gene encoding MORs) mRNA with glutamatergic (Vglut2) and neurokinin-1 receptor (Tacr1) mRNA in medullary and pontine regions involved in breathing control and modulation. We found that >50% of cells expressed Oprm1 mRNA in the preBötzinger complex (preBötC), nucleus tractus solitarius (NTS), nucleus ambiguus (NA), postinspiratory complex (PiCo), locus coeruleus (LC), Kölliker-Fuse nucleus (KF), and the lateral and medial parabrachial nuclei (LBPN and MPBN, respectively). Among Tacr1 mRNA-expressing cells, >50% coexpressed Oprm1 mRNA in the preBötC, NTS, NA, Bötzinger complex (BötC), PiCo, LC, raphe magnus nucleus, KF, LPBN, and MPBN, whereas among Vglut2 mRNA-expressing cells, >50% coexpressed Oprm1 mRNA in the preBötC, NTS, NA, BötC, PiCo, LC, KF, LPBN, and MPBN. Taken together, our study provides a comprehensive map of the distribution and coexpression of Oprm1, Tacr1, and Vglut2 mRNA in brainstem regions that control and modulate breathing and identifies Tacr1 and Vglut2 mRNA-expressing cells as subpopulations with potential vulnerability to modulation by opioid drugs.NEW & NOTEWORTHY Opioid drugs can cause serious respiratory side-effects by binding to µ-opioid receptors (MORs) in brainstem regions that control breathing. To better understand the regions and their cellular subpopulations that may be vulnerable to modulation by opioids, we provide a comprehensive map of Oprm1 (gene encoding MORs) mRNA expression throughout brainstem regions that control and modulate breathing. Notably, we identify glutamatergic and neurokinin-1 receptor-expressing cells as potentially vulnerable to modulation by opioid drugs and worthy of further investigation using targeted approaches.


Subject(s)
Receptors, Neurokinin-1 , Receptors, Opioid, mu , Vesicular Glutamate Transport Protein 2 , Animals , Receptors, Opioid, mu/metabolism , Receptors, Opioid, mu/genetics , Receptors, Neurokinin-1/metabolism , Receptors, Neurokinin-1/genetics , Mice , Vesicular Glutamate Transport Protein 2/metabolism , Vesicular Glutamate Transport Protein 2/genetics , Male , Brain Stem/metabolism , Brain Stem/drug effects , Mice, Inbred C57BL , RNA, Messenger/metabolism , RNA, Messenger/genetics , Respiratory Center/metabolism , Respiratory Center/drug effects
8.
Nature ; 629(8014): 1133-1141, 2024 May.
Article in English | MEDLINE | ID: mdl-38750368

ABSTRACT

The N-methyl-D-aspartate (NMDA) receptor is a glutamate-activated cation channel that is critical to many processes in the brain. Genome-wide association studies suggest that glutamatergic neurotransmission and NMDA receptor-mediated synaptic plasticity are important for body weight homeostasis1. Here we report the engineering and preclinical development of a bimodal molecule that integrates NMDA receptor antagonism with glucagon-like peptide-1 (GLP-1) receptor agonism to effectively reverse obesity, hyperglycaemia and dyslipidaemia in rodent models of metabolic disease. GLP-1-directed delivery of the NMDA receptor antagonist MK-801 affects neuroplasticity in the hypothalamus and brainstem. Importantly, targeting of MK-801 to GLP-1 receptor-expressing brain regions circumvents adverse physiological and behavioural effects associated with MK-801 monotherapy. In summary, our approach demonstrates the feasibility of using peptide-mediated targeting to achieve cell-specific ionotropic receptor modulation and highlights the therapeutic potential of unimolecular mixed GLP-1 receptor agonism and NMDA receptor antagonism for safe and effective obesity treatment.


Subject(s)
Dizocilpine Maleate , Glucagon-Like Peptide 1 , Glucagon-Like Peptide-1 Receptor , Obesity , Receptors, N-Methyl-D-Aspartate , Animals , Humans , Male , Mice , Rats , Brain Stem/metabolism , Brain Stem/drug effects , Disease Models, Animal , Dizocilpine Maleate/adverse effects , Dizocilpine Maleate/pharmacology , Dizocilpine Maleate/therapeutic use , Dyslipidemias/drug therapy , Dyslipidemias/metabolism , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Hyperglycemia/drug therapy , Hyperglycemia/metabolism , Hypothalamus/drug effects , Hypothalamus/metabolism , Mice, Inbred C57BL , Neuronal Plasticity/drug effects , Obesity/drug therapy , Obesity/metabolism , Rats, Sprague-Dawley , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors
9.
Epilepsia ; 65(7): e125-e130, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38738911

ABSTRACT

Because of its involvement in breathing control and neuronal excitability, dysregulation of the serotonin (5-HT) 2C receptor (5-HT2C) might play a key role in sudden unexpected death in epilepsy. Seizure-induced respiratory arrest is thus prevented by a 5-HT2B/C agonist in different seizure model. However, the specific contribution of 5-HT2C in chronic epilepsy-related respiratory dysfunction remains unknown. In a rat model of temporal lobe epilepsy (EPI rats), in which we previously reported interictal respiratory dysfunctions and a reduction of brainstem 5-HT tone, quantitative reverse transcriptase polymerase chain reaction showed overexpression of TPH2 (5-HT synthesis enzyme), SERT (5-HT reuptake transporter), and 5-HT2C transcript levels in the brainstem of EPI rats, and of RNA-specific adenosine deaminase (ADAR1, ADAR2) involved in the production of 5-HT2C isoforms. Interictal ventilation was assessed with whole-body plethysmography before and 2 h after administration of SB242084 (2 mg/kg), a specific antagonist of 5-HT2C. As expected, SB242084 administration induced a progressive decrease in ventilatory parameters and an alteration of breathing stability in both control and EPI rats. However, the size of the SB242084 effect was lower in EPI rats than in controls. Increased 5-HT2C gene expression in the brainstem of EPI rats could be part of a compensatory mechanism against epilepsy-related low 5-HT tone and expression of 5-HT2C isoforms for which 5-HT affinity might be lower.


Subject(s)
Brain Stem , Disease Models, Animal , Epilepsy, Temporal Lobe , Receptor, Serotonin, 5-HT2C , Animals , Receptor, Serotonin, 5-HT2C/genetics , Receptor, Serotonin, 5-HT2C/metabolism , Rats , Epilepsy, Temporal Lobe/physiopathology , Epilepsy, Temporal Lobe/metabolism , Brain Stem/metabolism , Brain Stem/drug effects , Male , Tryptophan Hydroxylase/genetics , Tryptophan Hydroxylase/metabolism , Serotonin Plasma Membrane Transport Proteins/genetics , Serotonin Plasma Membrane Transport Proteins/metabolism , Indoles/pharmacology , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Rats, Sprague-Dawley , Serotonin 5-HT2 Receptor Antagonists/pharmacology , Aminopyridines , Thiophenes
10.
Brain Res ; 1835: 148908, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38582416

ABSTRACT

BDNF, a neurotrophic factor, and its receptors have been implicated in the pathophysiology of mild traumatic brain injury (mTBI). The brainstem houses many vital functions, that are also associated with signs and symptoms of mTBI, but has been understudied in mTBI animal models. We determined the extent to which neurotrophic protein and associated receptor expression is affected within the brainstem of adult rats following mTBI. Their behavioral function was assessed and temporal expression of the 'negative' regulators of neuronal function (p75, t-TrkB, and pro-BDNF) and 'positive' neuroprotective (FL-TrkB and m-BDNF) protein isoforms were determined via western blot and immunohistochemistry at 1, 3, 7, and 14 post-injury days (PID) following mTBI or sham (control) procedure. Within the brainstem, p75 expression increased at PID 1 vs. sham animals. t-TrkB and pro-BDNF expression increased at PID 7 and 14. The 'positive' protein isoforms of FL-TrkB and m-BDNF expression were increased only at PID 7. The ratio of t-TrkB:FL-TrkB (negative:positive) was substantial across groups and time points, suggesting a negative impact of neurotrophic signaling on neuronal function. Additional NeuN experiments revealed cell death occurring within a subset of neurons within the medulla. While behavioral measures improved by PID 7-14, negative neurotrophic biochemical responses persisted. Despite the assertion that mTBI produces "mild" injury, evidence of cell death was observed in the medulla. Ratios of TrkB and BDNF isoforms with conflicting functions suggest that future work should specifically measure each subtype since they induce opposing downstream effects on neuronal function.


Subject(s)
Brain Stem , Brain-Derived Neurotrophic Factor , Rats, Sprague-Dawley , Receptor, trkB , Animals , Male , Brain-Derived Neurotrophic Factor/metabolism , Receptor, trkB/metabolism , Brain Stem/metabolism , Rats , Brain Concussion/metabolism , Disease Models, Animal , Neurons/metabolism , Protein Isoforms/metabolism , Time Factors , Nerve Tissue Proteins/metabolism , Brain Injuries, Traumatic/metabolism
11.
J Alzheimers Dis ; 99(1): 307-319, 2024.
Article in English | MEDLINE | ID: mdl-38669537

ABSTRACT

Background: Alzheimer's disease (AD) pathology is considered to begin in the brainstem, and cerebral microglia are known to play a critical role in AD pathogenesis, yet little is known about brainstem microglia in AD. Translocator protein (TSPO) PET, sensitive to activated microglia, shows high signal in dorsal brainstem in humans, but the precise location and clinical correlates of this signal are unknown. Objective: To define age and AD associations of brainstem TSPO PET signal in humans. Methods: We applied new probabilistic maps of brainstem nuclei to quantify PET-measured TSPO expression over the whole brain including brainstem in 71 subjects (43 controls scanned using 11C-PK11195; 20 controls and 8 AD subjects scanned using 11C-PBR28). We focused on inferior colliculi (IC) because of visually-obvious high signal in this region, and potential relevance to auditory dysfunction in AD. We also assessed bilateral cortex. Results: TSPO expression was normally high in IC and other brainstem regions. IC TSPO was decreased with aging (p = 0.001) and in AD subjects versus controls (p = 0.004). In cortex, TSPO expression was increased with aging (p = 0.030) and AD (p = 0.033). Conclusions: Decreased IC TSPO expression with aging and AD-an opposite pattern than in cortex-highlights underappreciated regional heterogeneity in microglia phenotype, and implicates IC in a biological explanation for strong links between hearing loss and AD. Unlike in cerebrum, where TSPO expression is considered pathological, activated microglia in IC and other brainstem nuclei may play a beneficial, homeostatic role. Additional study of brainstem microglia in aging and AD is needed.


Subject(s)
Aging , Alzheimer Disease , Brain Stem , Microglia , Positron-Emission Tomography , Receptors, GABA , Humans , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , Microglia/metabolism , Microglia/pathology , Male , Aged , Female , Aging/pathology , Brain Stem/metabolism , Brain Stem/pathology , Receptors, GABA/metabolism , Aged, 80 and over , Middle Aged , Isoquinolines , Adult
12.
Neuropathol Appl Neurobiol ; 50(3): e12977, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38680020

ABSTRACT

AIM: Leigh syndrome (LS), the most common paediatric presentation of genetic mitochondrial dysfunction, is a multi-system disorder characterised by severe neurologic and metabolic abnormalities. Symmetric, bilateral, progressive necrotizing lesions in the brainstem are defining features of the disease. Patients are often symptom free in early life but typically develop symptoms by about 2 years of age. The mechanisms underlying disease onset and progression in LS remain obscure. Recent studies have shown that the immune system causally drives disease in the Ndufs4(-/-) mouse model of LS: treatment of Ndufs4(-/-) mice with the macrophage-depleting Csf1r inhibitor pexidartinib prevents disease. While the precise mechanisms leading to immune activation and immune factors involved in disease progression have not yet been determined, interferon-gamma (IFNγ) and interferon gamma-induced protein 10 (IP10) were found to be significantly elevated in Ndufs4(-/-) brainstem, implicating these factors in disease. Here, we aimed to explore the role of IFNγ and IP10 in LS. METHODS: To establish the role of IFNγ and IP10 in LS, we generated IFNγ and IP10 deficient Ndufs4(-/-)/Ifng(-/-) and Ndufs4(-/-)/IP10(-/-) double knockout animals, as well as IFNγ and IP10 heterozygous, Ndufs4(-/-)/Ifng(+/-) and Ndufs4(-/-)/IP10(+/-), animals. We monitored disease onset and progression to define the impact of heterozygous or homozygous loss of IFNγ and IP10 in LS. RESULTS: Loss of IP10 does not significantly impact the onset or progression of disease in the Ndufs4(-/-) model. IFNγ loss significantly extends survival and delays disease progression in a gene dosage-dependent manner, though the benefits are modest compared to Csf1r inhibition. CONCLUSIONS: IFNγ contributes to disease onset and progression in LS. Our findings suggest that IFNγ targeting therapies may provide some benefits in genetic mitochondrial disease, but targeting IFNγ alone would likely yield only modest benefits in LS.


Subject(s)
Disease Progression , Electron Transport Complex I , Interferon-gamma , Leigh Disease , Animals , Mice , Brain Stem/pathology , Brain Stem/metabolism , Disease Models, Animal , Electron Transport Complex I/genetics , Electron Transport Complex I/deficiency , Interferon-gamma/metabolism , Leigh Disease/pathology , Leigh Disease/genetics , Mice, Inbred C57BL , Mice, Knockout
13.
Brain Struct Funct ; 229(5): 1121-1142, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38578351

ABSTRACT

In mammals, the ventral respiratory column (VRC) plays a pivotal role in integrating neurochemically diverse inputs from brainstem and forebrain regions to generate respiratory motor patterns. VRC microinjection of the neuropeptide galanin has been reported to dampen carbon dioxide (CO2)-mediated chemoreflex responses. Additionally, we previously demonstrated that galaninergic neurons in the retrotrapezoid nucleus (RTN) are implicated in the adaptive response to hypercapnic stimuli, suggesting a link between RTN neuroplasticity and increased neuronal drive to the VRC. VRC neurons express galanin receptor 1, suggesting potential regulatory action by galanin, however, the precise galaninergic chemoreceptor-VRC circuitry remains to be determined. This study aimed to identify sources of galaninergic input to the VRC that contribute to central respiratory chemoreception. We employed a combination of retrograde neuronal tracing, in situ hybridisation and immunohistochemistry to investigate VRC-projecting neurons that synthesise galanin mRNA. In an additional series of experiments, we used acute hypercapnia exposure (10% CO2, 1 h) and c-Fos immunohistochemistry to ascertain which galaninergic nuclei projecting to the VRC are activated. Our findings reveal that a total of 30 brain nuclei and 51 subnuclei project to the VRC, with 12 of these containing galaninergic neurons, including the RTN. Among these galaninergic populations, only a subset of the RTN neurons (approximately 55%) exhibited activation in response to acute hypercapnia. Our findings highlight that the RTN is the likely source of galaninergic transmission to the VRC in response to hypercapnic stimuli.


Subject(s)
Galanin , Hypercapnia , Neurons , Animals , Hypercapnia/metabolism , Hypercapnia/physiopathology , Male , Galanin/metabolism , Neurons/metabolism , Carbon Dioxide/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Neural Pathways/metabolism , Neural Pathways/physiology , Respiratory Center/metabolism , Rats , Chemoreceptor Cells/metabolism , Rats, Sprague-Dawley , Brain Stem/metabolism
14.
Am J Physiol Lung Cell Mol Physiol ; 326(6): L698-L712, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38591125

ABSTRACT

Chronic intermittent hypoxia (CIH) is a prevalent condition characterized by recurrent episodes of oxygen deprivation, linked to respiratory and neurological disorders. Prolonged CIH is known to have adverse effects, including endothelial dysfunction, chronic inflammation, oxidative stress, and impaired neuronal function. These factors can contribute to serious comorbidities, including metabolic disorders and cardiovascular diseases. To investigate the molecular impact of CIH, we examined male C57BL/6J mice exposed to CIH for 21 days, comparing with normoxic controls. We used single-nucleus RNA sequencing to comprehensively examine the transcriptomic impact of CIH on key cell classes within the brainstem, specifically excitatory neurons, inhibitory neurons, and oligodendrocytes. These cell classes regulate essential physiological functions, including autonomic tone, cardiovascular control, and respiration. Through analysis of 10,995 nuclei isolated from pontine-medullary tissue, we identified seven major cell classes, further subdivided into 24 clusters. Our findings among these cell classes, revealed significant differential gene expression, underscoring their distinct responses to CIH. Notably, neurons exhibited transcriptional dysregulation of genes associated with synaptic transmission, and structural remodeling. In addition, we found dysregulated genes encoding ion channels and inflammatory response. Concurrently, oligodendrocytes exhibited dysregulated genes associated with oxidative phosphorylation and oxidative stress. Utilizing CellChat network analysis, we uncovered CIH-dependent altered patterns of diffusible intercellular signaling. These insights offer a comprehensive transcriptomic cellular atlas of the pons-medulla and provide a fundamental resource for the analysis of molecular adaptations triggered by CIH.NEW & NOTEWORTHY This study on chronic intermittent hypoxia (CIH) from pons-medulla provides initial insights into the molecular effects on excitatory neurons, inhibitory neurons, and oligodendrocytes, highlighting our unbiased approach, in comparison with earlier studies focusing on single target genes. Our findings reveal that CIH affects cell classes distinctly, and the dysregulated genes in distinct cell classes are associated with synaptic transmission, ion channels, inflammation, oxidative stress, and intercellular signaling, advancing our understanding of CIH-induced molecular responses.


Subject(s)
Hypoxia , Mice, Inbred C57BL , Neurons , Oligodendroglia , Transcriptome , Animals , Oligodendroglia/metabolism , Mice , Male , Hypoxia/metabolism , Hypoxia/genetics , Neurons/metabolism , Neurons/pathology , Brain Stem/metabolism
15.
Brain Behav Immun ; 119: 333-350, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38561095

ABSTRACT

Neonatal sepsis remains one of the leading causes of mortality in newborns. Several brainstem-regulated physiological processes undergo disruption during neonatal sepsis. Mechanistic knowledge gaps exist at the interplay between metabolism and immune activation to brainstem neural circuits and pertinent physiological functions in neonates. To delineate this association, we induced systemic inflammation either by TLR4 (LPS) or TLR1/2 (PAM3CSK4) ligand administration in postnatal day 5 mice (PD5). Our findings show that LPS and PAM3CSK4 evoke substantial changes in respiration and metabolism. Physiological trade-offs led to hypometabolic-hypothermic responses due to LPS, but not PAM3CSK4, whereas to both TLR ligands blunted respiratory chemoreflexes. Neuroinflammatory pathways modulation in brainstem showed more robust effects in LPS than PAM3CSK4. Brainstem neurons, microglia, and astrocyte gene expression analyses showed unique responses to TLR ligands. PAM3CSK4 did not significantly modulate gene expression changes in GLAST-1 positive brainstem astrocytes. PD5 pups receiving PAM3CSK4 failed to maintain a prolonged metabolic state repression, which correlated to enhanced gasping latency and impaired autoresuscitation during anoxic chemoreflex challenges. In contrast, LPS administered pups showed no significant changes in anoxic chemoreflex. Electrophysiological studies from brainstem slices prepared from pups exposed to either TLR4 or PAM3CSK4 showed compromised transmission between preBötzinger complex and Hypoglossal as an exclusive response to the TLR1/2 ligand. Spatial gene expression analysis demonstrated a region-specific modulation of PAM3CSK4 within the raphe nucleus relative to other anatomical sites evaluated. Our findings suggest that metabolic changes due to inflammation might be a crucial tolerance mechanism for neonatal sepsis preserving neural control of breathing.


Subject(s)
Animals, Newborn , Brain Stem , Lipopolysaccharides , Neonatal Sepsis , Toll-Like Receptor 1 , Toll-Like Receptor 2 , Toll-Like Receptor 4 , Animals , Mice , Toll-Like Receptor 4/metabolism , Lipopolysaccharides/pharmacology , Toll-Like Receptor 2/metabolism , Neonatal Sepsis/metabolism , Brain Stem/metabolism , Toll-Like Receptor 1/metabolism , Lipopeptides/pharmacology , Respiration/drug effects , Mice, Inbred C57BL , Neurons/metabolism , Astrocytes/metabolism , Male , Ligands , Microglia/metabolism , Female , Inflammation/metabolism
16.
Brain Res ; 1836: 148938, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38615924

ABSTRACT

Prepulse inhibition (PPI) of the auditory startle response, a key measure of sensorimotor gating, diminishes with age and is impaired in various neurological conditions. While PPI deficits are often associated with cognitive impairments, their reversal is routinely used in experimental systems for antipsychotic drug screening. Yet, the cellular and circuit-level mechanisms of PPI remain unclear, even under non-pathological conditions. We recently showed that brainstem neurons located in the caudal pontine reticular nucleus (PnC) expressing the glycine transporter type 2 (GlyT2±) receive inputs from the central nucleus of the amygdala (CeA) and contribute to PPI but via an uncharted pathway. Here, using tract-tracing, immunohistochemistry and in vitro optogenetic manipulations coupled to field electrophysiological recordings, we reveal the neuroanatomical distribution of GlyT2± PnC neurons and PnC-projecting CeA glutamatergic neurons and we provide mechanistic insights on how these glutamatergic inputs suppress auditory neurotransmission in PnC sections. Additionally, in vivo experiments using GlyT2-Cre mice confirm that optogenetic activation of GlyT2± PnC neurons enhances PPI and is sufficient to induce PPI in young mice, emphasizing their role. However, in older mice, PPI decline is not further influenced by inhibiting GlyT2± neurons. This study highlights the importance of GlyT2± PnC neurons in PPI and underscores their diminished activity in age-related PPI decline.


Subject(s)
Brain Stem , Glycine Plasma Membrane Transport Proteins , Glycine , Neurons , Prepulse Inhibition , Reflex, Startle , Animals , Prepulse Inhibition/physiology , Neurons/physiology , Neurons/metabolism , Reflex, Startle/physiology , Mice , Brain Stem/physiology , Brain Stem/metabolism , Glycine Plasma Membrane Transport Proteins/metabolism , Male , Glycine/metabolism , Optogenetics , Mice, Transgenic , Mice, Inbred C57BL , Synaptic Transmission/physiology , Central Amygdaloid Nucleus/physiology , Central Amygdaloid Nucleus/metabolism
17.
J Parkinsons Dis ; 14(4): 823-831, 2024.
Article in English | MEDLINE | ID: mdl-38640171

ABSTRACT

Background: Rapid eye movement sleep behavior disorder (RBD) may precede or follow motor symptoms in Parkinson's disease (PD). While over 70% of idiopathic RBD cases phenoconvert within a decade, a small subset develops PD after a more extended period or remains nonconverted. These heterogeneous manifestations of RBD in PD prompt subtype investigations. Premotor RBD may signify "body-first" PD with bottom-up, symmetric synucleinopathy propagation. Objective: Explore brainstem and nigrostriatal monoaminergic degeneration pattern differences based on premotor RBD presence and duration in de novo PD patients. Methods: In a cross-sectional analysis of de novo PD patients (n = 150) undergoing FP-CIT PET and RBD Single-Question Screen, the cohort was categorized into groups with and without premotor RBD (PDRBD +/-), with further classification of PDRBD + based on a 10-year duration of premotor RBD. Analysis of FP-CIT binding in the striatum and pons, striatal asymmetry, and striatum-to-pons ratios compared patterns of nigrostriatal and brainstem monoaminergic degeneration. Results: PDRBD + exhibited more severe and symmetrical striatal dopaminergic denervation compared to PDRBD-, with the difference in severity accentuated in the least-affected hemisphere. The PDRBD +<10Y subgroup displayed the most prominent striatal symmetry, supporting a more homogeneous "body-first" subtype. Pontine uptakes remained lower in PDRBD + even after adjusting for striatal uptake, suggesting early degeneration of pontine monoaminergic nuclei. Conclusions: Premotor RBD in PD is associated with severe, symmetrical nigrostriatal and brainstem monoaminergic degeneration, especially in cases with PD onset within 10 years of RBD. This supports the concept of a "widespread, bottom-up" pathophysiological mechanism associated with premotor RBD in PD.


Subject(s)
Parkinson Disease , Positron-Emission Tomography , REM Sleep Behavior Disorder , Humans , REM Sleep Behavior Disorder/metabolism , REM Sleep Behavior Disorder/etiology , REM Sleep Behavior Disorder/diagnostic imaging , REM Sleep Behavior Disorder/pathology , Parkinson Disease/complications , Parkinson Disease/metabolism , Parkinson Disease/diagnostic imaging , Parkinson Disease/pathology , Parkinson Disease/physiopathology , Male , Aged , Female , Middle Aged , Cross-Sectional Studies , Corpus Striatum/metabolism , Corpus Striatum/diagnostic imaging , Corpus Striatum/pathology , Brain Stem/diagnostic imaging , Brain Stem/metabolism , Brain Stem/pathology , Tropanes , Substantia Nigra/diagnostic imaging , Substantia Nigra/metabolism , Substantia Nigra/pathology
18.
Synapse ; 78(2): e22289, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38436644

ABSTRACT

Epileptic seizures are seen as a result of changing excitability balance depending on the deterioration in synaptic plasticity in the brain. Neuroplastin, and its related molecules which are known to play a role in synaptic plasticity, neurotransmitter activities that provide balance of excitability and, different neurological diseases, have not been studied before in epilepsy. In this study, a total of 34 Sprague-Dawley male and female rats, 2 months old, weighing 250-300 g were used. The epilepsy model in rats was made via pentylenetetrazole (PTZ). After the completion of the experimental procedure, the brain tissue of the rats were taken and the histopathological changes in the hippocampus and cortex parts and the brain stem were investigated, as well as the immunoreactivity of the proteins related to the immunohistochemical methods. As a result of the histopathological evaluation, it was determined that neuron degeneration and the number of dilated blood vessels in the hippocampus, frontal cortex, and brain stem were higher in the PTZ status epilepticus (SE) groups than in the control groups. It was observed that neuroplastin and related proteins TNF receptor-associated factor 6 (TRAF6), Gamma amino butyric acid type A receptors [(GABA(A)], and plasma membrane Ca2+ ATPase (PMCA) protein immunoreactivity levels increased especially in the male hippocampus, and only AMPA receptor subunit type 1 (GluA1) immunoreactivity decreased, unlike other proteins. We believe this may be caused by a problem in the mechanisms regulating the interaction of neuroplastin and GluA1 and may cause problems in synaptic plasticity in the experimental epilepsy model. It may be useful to elucidate this mechanism and target GluA1 when determining treatment strategies.


Subject(s)
Epilepsy , Animals , Female , Male , Rats , Brain Stem/metabolism , Epilepsy/chemically induced , Epilepsy/genetics , Hippocampus/metabolism , Pentylenetetrazole , Rats, Sprague-Dawley , Receptors, GABA-A/genetics , TNF Receptor-Associated Factor 6/genetics , Plasma Membrane Calcium-Transporting ATPases/genetics , Receptors, AMPA/genetics , Cerebral Cortex/metabolism
19.
J Neurochem ; 168(5): 663-676, 2024 May.
Article in English | MEDLINE | ID: mdl-38439211

ABSTRACT

Neurons exhibit a high energetic need, and the question arises as how they metabolically adapt to changing activity states. This is relevant for interpreting functional neuroimaging in different brain areas. Particularly, neurons with a broad firing range might exhibit metabolic adaptations. Therefore, we studied MNTB (medial nucleus of the trapezoid body) principal neurons, which generate action potentials (APs) at frequencies up to several hundred hertz. We performed the experiments in acute brainstem slices of the Mongolian gerbil (Meriones unguiculatus) at 22.5-24.5°C. Upon electrical stimulation of afferent MNTB fibres with 400 stimuli at varying frequencies, we monitored autofluorescence levels of NAD(P)H and FAD and determined the extremum amplitudes of their biphasic response. Additionally, we compared these data with alterations in O2 concentrations measured with an electrochemical sensor. These O2 changes are prominent since MNTB neurons rely on oxidative phosphorylation as shown by our pharmacological experiments. We calculated the O2 consumption rate as change in O2 concentration divided by stimulus durations, because these periods varied inversely with stimulus frequency as a result of the constant number of 400 stimuli applied. The O2 consumption rate increased with stimulation frequency up to a constant value at 600 Hz; that is, energy demand depends on temporal characteristics of activity despite the same number of stimuli. The rates showed no correlation with peak amplitudes of NAD(P)H or FAD, whilst the values of the two molecules were linearly correlated. This points at the complexity of analysing autofluorescence imaging for quantitative metabolic studies, because these values report only relative net changes of many superimposed oxidative and reductive processes. Monitoring O2 concentration rates is, thus, an important tool to improve the interpretation of NAD(P)H/FAD autofluorescence data, as they do not under all conditions and in all systems appropriately reflect the metabolic activity or energy demand.


Subject(s)
Brain Stem , Gerbillinae , Neurons , Animals , Neurons/metabolism , Neurons/physiology , Brain Stem/metabolism , Oxygen Consumption/physiology , Action Potentials/physiology , Male , Electric Stimulation , Flavin-Adenine Dinucleotide/metabolism , Female , Trapezoid Body/physiology , Trapezoid Body/metabolism , NADP/metabolism
20.
Curr Biol ; 34(8): 1646-1656.e4, 2024 04 22.
Article in English | MEDLINE | ID: mdl-38518777

ABSTRACT

The obesity epidemic is principally driven by the consumption of more calories than the body requires. It is therefore essential that the mechanisms underpinning feeding behavior are defined. Neurons within the brainstem dorsal vagal complex (DVC) receive direct information from the digestive system and project to second-order regions in the brain to regulate food intake. Although γ-aminobutyric acid is expressed in the DVC (GABADVC), its function in this region has not been defined. In order to discover the unique gene expression signature of GABADVC cells, we used single-nucleus RNA sequencing (Nuc-seq), and this revealed 19 separate clusters. We next probed the function of GABADVC cells and discovered that the selective activation of GABADVC neurons significantly controls food intake and body weight. Optogenetic interrogation of GABADVC circuitry identified GABADVC → hypothalamic arcuate nucleus (ARC) projections as appetite suppressive without creating aversion. Electrophysiological analysis revealed that GABADVC → ARC stimulation inhibits hunger-promoting neuropeptide Y (NPY) neurons via GABA release. Adopting an intersectional genetics strategy, we clarify that the GABADVC → ARC circuit curbs food intake. These data identify GABADVC as a new modulator of feeding behavior and body weight and a controller of orexigenic NPY neuron activity, thereby providing insight into the neural underpinnings of obesity.


Subject(s)
Arcuate Nucleus of Hypothalamus , Brain Stem , Feeding Behavior , Arcuate Nucleus of Hypothalamus/metabolism , Arcuate Nucleus of Hypothalamus/physiology , Animals , Brain Stem/physiology , Brain Stem/metabolism , Mice , Male , Feeding Behavior/physiology , GABAergic Neurons/physiology , GABAergic Neurons/metabolism , gamma-Aminobutyric Acid/metabolism , Eating/physiology , Mice, Inbred C57BL , Female
SELECTION OF CITATIONS
SEARCH DETAIL
...