Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Brain Sci ; 13(2)2023 Feb 03.
Article in English | MEDLINE | ID: mdl-36831801

ABSTRACT

Huntingtin-associated protein 1 (HAP1) is a determinant marker for the stigmoid body (STB), a neurocytoplasmic physiological inclusion. STB/HAP1 enriched areas in the brain/spinal cord are usually protected from neurodegenerative diseases, whereas the regions with tiny amounts or no STB/HAP1 are affected. In addition to the brain/spinal cord, HAP1 is highly expressed in the myenteric/submucosal plexuses of the enteric nervous system in the gastrointestinal tract. The tongue is attached to the pharynx by the hyoid bone as an extension of the gastrointestinal system. To date, the immunohistochemical distribution and neurochemical characterization of HAP1 have not been elucidated in the lingual ganglia. Using immunohistochemistry and light microscopy, our current study demonstrates the expression and immunohistochemical phenotype of HAP1 in the lingual ganglia of adult mice. We showed that HAP1 was profoundly distributed in the intralingual ganglion (ILG) and the ganglia near the root of the tongue (which we coined as "lingual root ganglion"; LRG). Neurons in ILG and LRG exhibited high coexpression of HAP1 with NOS or ChAT. Furthermore, most HAP1-immunoreactive neurons contained SP, CGRP, and VIP immunoreactivity in both ILG and LRG. The current results might serve as an essential base for future studies to elucidate the pathological/physiological functions of HAP1 in the lingual ganglia.

2.
Neurosci Res ; 191: 13-27, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36581175

ABSTRACT

Huntingtin-associated protein 1(HAP1) is an immunohistochemical marker of the stigmoid body (STB). Brain and spinal cord regions with lack of STB/HAP1 immunoreactivity are always neurodegenerative targets, whereas STB/HAP1 abundant regions are usually spared from neurodegeneration. In addition to the brain and spinal cord, HAP1 is abundantly expressed in the excitatory and inhibitory motor neurons in myenteric plexuses of the enteric nervous system (ENS). However, the detailed expression of HAP1 and its neurochemical characterization in submucosal plexuses of ENS are still unknown. In this study, we aimed to clarify the expression and neurochemical characterization of HAP1 in the submucosal plexuses of the small intestine in adult mice and rats. HAP1 was highly expressed in the submucosal plexuses of both rodents. The percentage of HAP1-immunoreactive submucosal neurons was not significantly varied between the intestinal segments of these rodents. Double immunofluorescence results revealed that almost all the cholinergic secretomotor neurons containing ChAT/ CGRP/ somatostatin/ calretinin, non-cholinergic secretomotor neurons containing VIP/NOS/TH/calretinin, and vasodilator neurons containing VIP/calretinin expressed HAP1. Our current study is the first to clarify that STB/HAP1 is expressed in secretomotor and vasodilator neurons of submucosal plexuses, suggesting that STB/HAP1 might modulate or protect the secretomotor and vasodilator functions of submucosal neurons in ENS.


Subject(s)
Rodentia , Vasodilator Agents , Rats , Mice , Animals , Calbindin 2/metabolism , Vasodilator Agents/metabolism , Intestine, Small , Myenteric Plexus/metabolism , Motor Neurons , Phenotype
3.
Brain Res ; 1798: 148129, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36332665

ABSTRACT

The localization and function of synaptotagmin (syt)17 in the suprachiasmatic nucleus (SCN) of the brain, which is the master circadian oscillator, were investigated. The Syt17 mRNA-containing neurons were mainly situated in the shell region while SYT17 immunoreactive cell bodies and neural fibers were detected in the core and shell of the SCN and the subparaventricular zone (SPZ). Further, electron microscopy analysis revealed SYT17 in the rough endoplasmic reticulum (rER), Golgi apparatus (G), and large and small vesicles of neurons. Syt17 mRNA expression in the SCN showed a circadian rhythm, and light exposure at night suppressed its expression. In addition, the free running period of locomotor activity rhythm was shortened in Syt17-deletion mutant mice. These findings suggest that SYT17 is involved in the regulation of circadian rhythms.


Subject(s)
Circadian Rhythm , Suprachiasmatic Nucleus , Synaptotagmins , Animals , Mice , Circadian Rhythm/physiology , Mammals/genetics , Neurons/metabolism , RNA, Messenger/metabolism , Suprachiasmatic Nucleus/metabolism , Synaptotagmins/metabolism
4.
Neuroscience ; 499: 40-63, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35870563

ABSTRACT

Huntingtin-associated protein 1 (HAP1) is a core component of stigmoid body (STB) and is known as a neuroprotective interactor with causal agents for various neurodegenerative diseases. Brain regions rich in STB/HAP1 immunoreactivity are usually spared from cell death, whereas brain regions with negligible STB/HAP1 immunoreactivity are the major neurodegenerative targets. Recently, we have shown that STB/HAP1 is abundantly expressed in the spinal preganglionic sympathetic/parasympathetic neurons but absent in the motoneurons of spinal cord, indicating that spinal motoneurons are more vulnerable to neurodegenerative diseases. In light of STB/HAP1 neuroprotective effects, it is also essential to clarify the distribution of STB/HAP1 in another major neurodegenerative target, the brainstem. Here, we examined the expression and detailed immunohistochemical distribution of STB/HAP1 and its relationships with choline acetyltransferase (ChAT) in the midbrain, pons, and medulla oblongata of adult mice. Abundant STB/HAP1 immunoreactive neurons were disseminated in the periaqueductal gray, Edinger-Westphal nucleus, raphe nuclei, locus coeruleus, pedunculopontine tegmental nucleus, superior/inferior salivatory nucleus, and dorsal motor nucleus of vagus. Double-label immunohistochemistry of HAP1 with ChAT (or with urocortin-1 for Edinger-Westphal nucleus centrally projecting population) confirmed that STB/HAP1 was highly present in parasympathetic preganglionic neurons but utterly absent in cranial nerve motor nuclei throughout the brainstem. These results suggest that due to deficient putative STB/HAP1-protectivity, cranial nerve motor nuclei might be more vulnerable to certain neurodegenerative stresses than STB/HAP1-expressing brainstem nuclei, including preganglionic parasympathetic nuclei. Our current results also lay a basic foundation for future studies that seek to clarify the physiological/pathological roles of STB/HAP1 in the brainstem.


Subject(s)
Brain Stem , Choline O-Acetyltransferase , Animals , Brain Stem/metabolism , Choline O-Acetyltransferase/metabolism , Cranial Nerves/metabolism , Medulla Oblongata , Mice , Motor Neurons/metabolism , Nerve Tissue Proteins/metabolism
5.
Cell Tissue Res ; 386(3): 533-558, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34665322

ABSTRACT

Huntingtin-associated protein 1 (HAP1) is a neural huntingtin interactor and being considered as a core molecule of stigmoid body (STB). Brain/spinal cord regions with abundant STB/HAP1 expression are usually spared from neurodegeneration in stress/disease conditions, whereas the regions with little STB/HAP1 expression are always neurodegenerative targets. The enteric nervous system (ENS) can act as a potential portal for pathogenesis of neurodegenerative disorders. However, ENS is also a neurodegenerative target in these disorders. To date, the expression of HAP1 and its neurochemical characterization have never been examined there. In the current study, we determined the expression of HAP1 in the ENS of adult mice and characterized the morphological relationships of HAP1-immunoreactive (ir) cells with the markers of motor neurons, sensory neurons, and interneurons in the myenteric plexus using Western blotting and light/fluorescence microscopy. HAP1-immunoreaction was present in both myenteric and submucosal plexuses of ENS. Most of the HAP1-ir neurons exhibited STB in their cytoplasm. In myenteric plexus, a large number of calretinin, calbindin, NOS, VIP, ChAT, SP, somatostatin, and TH-ir neurons showed HAP1-immunoreactivity. In contrast, most of the CGRP-ir neurons were devoid of HAP1-immunoreactivity. Our current study is the first to clarify that HAP1 is highly expressed in excitatory motor neurons, inhibitory motor neurons, and interneurons but almost absent in sensory neurons in myenteric plexus. These suggest that STB/HAP1-ir neurons are mostly Dogiel type I neurons. Due to lack of putative STB/HAP1 protectivity, the sensory neurons (Dogiel type II) might be more vulnerable to neurodegeneration than STB/HAP1-expressing motoneurons/interneurons (Dogiel type I) in myenteric plexus.


Subject(s)
DNA-Binding Proteins/metabolism , Gastrointestinal Tract/physiology , Immunohistochemistry/methods , Myenteric Plexus/metabolism , Animals , Male , Mice , Phenotype
6.
Brain Sci ; 11(2)2021 Feb 14.
Article in English | MEDLINE | ID: mdl-33672867

ABSTRACT

Adolescence is the critical postnatal stage for the action of androgen in multiple brain regions. Androgens can regulate the learning/memory functions in the brain. It is known that the inhibitory avoidance test can evaluate emotional memory and is believed to be dependent largely on the amygdala and hippocampus. However, the effects of androgen on inhibitory avoidance memory have never been reported in adolescent male rats. In the present study, the effects of androgen on inhibitory avoidance memory and on androgen receptor (AR)-immunoreactivity in the amygdala and hippocampus were studied using behavioral analysis, Western blotting and immunohistochemistry in sham-operated, orchiectomized, orchiectomized + testosterone or orchiectomized + dihydrotestosterone-administered male adolescent rats. Orchiectomized rats showed significantly reduced time spent in the illuminated box after 30 min (test 1) or 24 h (test 2) of electrical foot-shock (training) and reduced AR-immunoreactivity in amygdala/hippocampal cornu Ammonis (CA1) in comparison to those in sham-operated rats. Treatment of orchiectomized rats with either non-aromatizable dihydrotestosterone or aromatizable testosterone were successfully reinstated these effects. Application of flutamide (AR-antagonist) in intact adolescent rats exhibited identical changes to those in orchiectomized rats. These suggest that androgens enhance the inhibitory avoidance memory plausibly by binding with AR in the amygdala and hippocampus.

7.
Acta Histochem ; 122(8): 151650, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33161374

ABSTRACT

Huntingtin-associated protein 1 (HAP1) is a neuronal cytoplasmic protein that is predominantly expressed in the brain and spinal cord. In addition to the central nervous system, HAP1 is also expressed in the peripheral organs including endocrine system. Different types of enteroendocrine cells (EEC) are present in the digestive organs. To date, the characterization of HAP1-immunoreactive (ir) cells remains unreported there. In the present study, the expression of HAP1 in pyloric stomach in adult male rats and its relationships with different chemical markers for EEC [gastrin, marker of gastrin (G) cells; somatostatin, marker of delta (D) cells; 5-HT, marker of enterochromaffin (EC) cells; histamine, marker of enterochromaffin-like (ECL) cells] were examined employing single- or double-labelled immunohistochemistry and with light-, fluorescence- or electron-microscopy. HAP1-ir cells were abundantly expressed in the glandular mucosa but were very few or none in the surface epithelium. Double-labelled immunofluorescence staining for HAP1 and markers for EECs showed that almost all the G-cells expressed HAP1. In contrast, HAP1 was completely lacking in D-cells, EC-cells or ECL-cells. Our current study is the first to clarify that HAP1 is selectively expressed in G-cells in rat pyloric stomach, which probably reflects HAP1's involvement in regulation of the secretion of gastrin.


Subject(s)
Enterochromaffin Cells/metabolism , Enterochromaffin-like Cells/metabolism , Gastric Mucosa/metabolism , Nerve Tissue Proteins/genetics , Pylorus/metabolism , Somatostatin-Secreting Cells/metabolism , Animals , Biomarkers/metabolism , Enterochromaffin Cells/cytology , Enterochromaffin-like Cells/cytology , Gastric Mucosa/cytology , Gastrins/biosynthesis , Gene Expression , Histamine/biosynthesis , Immunohistochemistry , Male , Nerve Tissue Proteins/metabolism , Organ Specificity , Pylorus/cytology , Rats , Rats, Wistar , Somatostatin/biosynthesis , Somatostatin-Secreting Cells/cytology
8.
IBRO Rep ; 9: 258-269, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33089002

ABSTRACT

Huntingtin-associated protein 1 (HAP1) is a polyglutamine (polyQ) length-dependent interactor with causal agents in several neurodegenerative diseases and has been regarded as a protective factor against neurodegeneration. In normal rodent brain and spinal cord, HAP1 is abundantly expressed in the areas that are spared from neurodegeneration while those areas with little HAP1 are frequent targets of neurodegeneration. We have recently showed that HAP1 is highly expressed in the spinal dorsal horn and may participate in modification/protection of certain sensory functions. Neurons in the dorsal root ganglia (DRG) transmits sensory stimuli from periphery to spinal cord/brain stem. Nevertheless, to date HAP1 expression in DRG remains unreported. In this study, the expression of HAP1 in cervical, thoracic, lumbar and sacral DRG in adult male mice and its relationships with different chemical markers for sensory neurons were examined using Western blot and immunohistochemistry. HAP1-immunoreactivity was detected in the cytoplasm of DRG neurons, and the percentage of HAP1-immunoreactive (ir) DRG neurons was ranged between 28-31 %. HAP1-immunoreactivity was comparatively more in the small cells (47-58 %) and medium cells (40-44 %) than that in the large cells (9-11 %). Double-immunostaining for HAP1 and markers for nociceptive or mechanoreceptive neurons showed that about 70-80 % of CGRP-, SP-, CB-, NOS-, TRPV1-, CR- and PV-ir neurons expressed HAP1. In contrast, HAP1 was completely lacking in TH-ir neurons. Our current study is the first to clarify that HAP1 is highly expressed in nociceptive/proprioceptive neurons but absent in light-touch-sensitive TH neurons, suggesting the potential importance of HAP1 in pain transduction and proprioception.

9.
Neuroscience ; 440: 15-29, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32450298

ABSTRACT

Androgen receptor (AR) is abundantly expressed in the preoptico-hypothalamic area, bed nucleus of stria terminalis, and medial amygdala of the brain where androgen plays an important role in regulating male sociosexual, emotional and aggressive behaviors. In addition to these brain regions, AR is also highly expressed in the hippocampus, suggesting that the hippocampus is another major target of androgenic modulation. It is known that androgen can modulate synaptic plasticity in the CA1 hippocampal subfield. However, to date, the effects of androgen on the intrinsic plasticity of hippocampal neurons have not been clearly elucidated. In this study, the effects of androgen on the expression of AR in the hippocampus and on the dynamics of intrinsic plasticity of CA1 pyramidal neurons were examined using immunohistochemistry, Western blotting and whole-cell current-clamp recording in unoperated, sham-operated, orchiectomized (OCX), OCX + testosterone (T) or OCX + dihydrotestosterone (DHT)-primed adolescent male rats. Orchiectomy significantly decreased AR-immunoreactivity, resting membrane potential, action potential numbers, afterhyperpolarization amplitude and membrane resistance, whereas it significantly increased action potential threshold and membrane capacitance. These effects were successfully reversed by treatment with either aromatizable androgen T or non-aromatizable androgen DHT. Furthermore, administration of the AR-antagonist flutamide in intact rats showed similar changes to those in OCX rats, suggesting that androgens affect the excitability of CA1 pyramidal neurons possibly by acting on the AR. Our current study potentially clarifies the role of androgen in enhancing the basal excitability of the CA1 pyramidal neurons, which may influence selective neuronal excitation/activation to modulate certain hippocampal functions.


Subject(s)
Androgens , Hippocampus , Androgens/pharmacology , Animals , Dihydrotestosterone/pharmacology , Flutamide/pharmacology , Hippocampus/metabolism , Male , Pyramidal Cells/metabolism , Rats , Receptors, Androgen/metabolism
10.
Neuroscience ; 394: 109-126, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30367943

ABSTRACT

Huntingtin-associated protein 1 (HAP1) is a neural interactor of huntingtin in Huntington's disease and interacts with gene products in a number of other neurodegenerative diseases. In normal brains, HAP1 is expressed abundantly in the hypothalamus and limbic-associated regions. These areas tend to be spared from neurodegeneration while those with little HAP1 are frequently neurodegenerative targets, suggesting its role as a protective factor against apoptosis. In light of the relationship between neurodegenerative diseases and deterioration of higher nervous activity, it is important to definitively clarify HAP1 expression in a cognitively important brain region, the retrosplenial-retrohippocampal area. Here, HAP1 expression was evaluated immunohistochemically over the retrosplenial cortex, the subicular complex, and the entorhinal and perirhinal cortices. HAP1-immunoreactive (ir) cells were classified into five discrete groups: (1) a distinct retrosplenial cell cluster exclusive to the superficial layers of the granular cortex, (2) a conspicuous, thin line of cells in layers IV/V of the "subiculum-backing cortex," (3) a group of highly immunoreactive cells associated with the medial entorhinal-subicular corner, (4) pericallosal cells just below layer VI and adjacent to the white matter, and (5) other sporadic, widely-disseminated HAP1-immunoreactive cells. HAP1 was found to be the first marker for the complex subiculum-backing cortex and a precise marker for several subfields in the retrosplenial-retrohippocampal area, verified through comparative staining with other neurochemicals. HAP1 may play an important role in protecting these cortical structures and functions for higher nervous activity by increasing the threshold to neurodegeneration and decreasing vulnerability to stress or aging.


Subject(s)
Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Hippocampus/cytology , Hippocampus/metabolism , Nerve Tissue Proteins/analysis , Animals , Corpus Callosum/cytology , Corpus Callosum/metabolism , Immunohistochemistry , Male , Neurons/cytology , Neurons/metabolism , Rats, Wistar
11.
Cell Rep ; 24(9): 2231-2247.e7, 2018 08 28.
Article in English | MEDLINE | ID: mdl-30157420

ABSTRACT

Sleep regulation involves interdependent signaling among specialized neurons in distributed brain regions. Although acetylcholine promotes wakefulness and rapid eye movement (REM) sleep, it is unclear whether the cholinergic pathway is essential (i.e., absolutely required) for REM sleep because of redundancy from neural circuits to molecules. First, we demonstrate that synaptic inhibition of TrkA+ cholinergic neurons causes a severe short-sleep phenotype and that sleep reduction is mostly attributable to a shortened sleep duration in the dark phase. Subsequent comprehensive knockout of acetylcholine receptor genes by the triple-target CRISPR method reveals that a similar short-sleep phenotype appears in the knockout of two Gq-type acetylcholine receptors Chrm1 and Chrm3. Strikingly, Chrm1 and Chrm3 double knockout chronically diminishes REM sleep to an almost undetectable level. These results suggest that muscarinic acetylcholine receptors, Chrm1 and Chrm3, are essential for REM sleep.


Subject(s)
Acetylcholine/metabolism , Receptor, Muscarinic M1/metabolism , Receptor, Muscarinic M3/metabolism , Sleep, REM/genetics , Animals , HEK293 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout
12.
Sci Rep ; 8(1): 854, 2018 01 16.
Article in English | MEDLINE | ID: mdl-29339832

ABSTRACT

In mammals, the principal circadian oscillator exists in the hypothalamic suprachiasmatic nucleus (SCN). In the SCN, CLOCK works as an essential component of molecular circadian oscillation, and ClockΔ19 mutant mice show unique characteristics of circadian rhythms such as extended free running periods, amplitude attenuation, and high-magnitude phase-resetting responses. Here we investigated what modifications occur in the spatiotemporal organization of clock gene expression in the SCN of ClockΔ19 mutants. The cultured SCN, sampled from neonatal homozygous ClockΔ19 mice on an ICR strain comprising PERIOD2::LUCIFERASE, demonstrated that the Clock gene mutation not only extends the circadian period, but also affects the spatial phase and period distribution of circadian oscillations in the SCN. In addition, disruption of the synchronization among neurons markedly attenuated the amplitude of the circadian rhythm of individual oscillating neurons in the mutant SCN. Further, with numerical simulations based on the present studies, the findings suggested that, in the SCN of the ClockΔ19 mutant mice, stable oscillation was preserved by the interaction among oscillating neurons, and that the orderly phase and period distribution that makes a phase wave are dependent on the functionality of CLOCK.


Subject(s)
CLOCK Proteins/metabolism , Neurons/metabolism , Suprachiasmatic Nucleus/metabolism , Animals , Behavior, Animal , CLOCK Proteins/genetics , Circadian Rhythm/physiology , Cyclic AMP/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Models, Theoretical , Mutagenesis , Period Circadian Proteins/genetics , Period Circadian Proteins/metabolism , Signal Transduction/drug effects , Suprachiasmatic Nucleus/cytology , Suprachiasmatic Nucleus/drug effects , Tetrodotoxin/pharmacology
13.
Acta Histochem Cytochem ; 50(2): 95-104, 2017 Apr 27.
Article in English | MEDLINE | ID: mdl-28522884

ABSTRACT

Both prokineticin receptor 2 (pkr2) and prokineticin 2 (pk2) gene-deficient mice have hypoplasia of the main olfactory bulb (MOB). This hypoplasia has been attributed to disruption of the glomerulus that is caused by loss of afferent projection from olfactory sensory neurons (OSN), and to the impaired migration of granule cells, a type of interneuron. In the present study, we examined whether migration of the second type of interneuron, periglomerular cells (PGC), is dependent on the pkr2 expression by observing the localization of distinct subpopulations of PGC: calretinin (CR)-, calbindin (CB)- and tyrosine hydroxylase (TH)-expressing neurons. In the Pkr2-/- mice, the construction of the layered structure of the MOB was partially preserved, with the exception of the internal plexiform layer (IPL) and the glomerular layer (GL). In the outermost layer of the MOB, abundant CR- and CB-immunopositive neurons were observed in the hypoplastic olfactory bulb. In addition, although markedly decreased, TH-immunopositive neurons were also observed in the outermost cell-dense region in the Pkr2-/-. The findings suggest that the migration of PGC to the MOB, as well as the migration from the core to the surface region of the MOB, is not driven by the PK2-PKR2 system.

14.
Zoolog Sci ; 30(12): 1011-8, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24320178

ABSTRACT

The mammalian circadian oscillator is composed of interacting positive and negative transcription events. The clock proteins PER1 and PER2 play essential roles in a negative limb of the feedback loop that generates the circadian rhythm in mammals. In addition, the proteins CLOCK and BMAL1 (also known as ARNTL) form a heterodimer that drives the Per genes via the E-box consensus sequences within their promoter regions. In the present study, we demonstrate that Id2 is involved in stabilization of the amplitudes of the circadian oscillations by suppressing transcriptional activation of clock genes Clock and Bmal1. Id2 shows dynamic oscillation in the SCN, with a peak in the late subjective night. Under constant dark conditions (DD), Id2(-/-) mice showed no apparent difference in locomotor activity, however, under constant light conditions (LL), Id2(-/-) mice exhibit aberrant locomotor activity, with lower circadian oscillation amplitudes, although the free running periods in Id2(-/-) mice show no differences from those in either wild type or heterozygous mice. Id2(-/-) animals also exhibit upregulation of Per1 in constant light, during both the subjective night and day. In wild type mice, Id2 is upregulated by constant light exposure during the subjective night. We propose that Id2 expression in the SCN contributes to maintenance of dynamic circadian oscillations.


Subject(s)
Circadian Rhythm/physiology , Gene Expression Regulation/physiology , Inhibitor of Differentiation Protein 2/metabolism , Light , Animals , CLOCK Proteins/genetics , CLOCK Proteins/metabolism , Cell Line , Female , Inhibitor of Differentiation Protein 2/genetics , Mice , Motor Activity
15.
Eur J Neurosci ; 38(6): 2832-41, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23869693

ABSTRACT

The suprachiasmatic nucleus (SCN) is the mammalian circadian rhythm center. Individual oscillating neurons have different endogenous circadian periods, but they are usually synchronized by an intercellular coupling mechanism. The differences in the period of each oscillating neuron have been extensively studied; however, the clustering of oscillators with similar periods has not been reported. In the present study, we artificially disrupted the intercellular coupling among oscillating neurons in the SCN and observed regional differences in the periods of the oscillating small-latticed regions of the SCN using a transgenic rat carrying a luciferase reporter gene driven by regulatory elements from a per2 clock gene (Per2::dluc rat). The analysis divided the SCN into two regions--aregion with periods shorter than 24 h (short-period region, SPR) and another with periods longer than 24 h (long-period region, LPR). The SPR was located in the smaller medial region of the dorsal SCN, whereas the LPR occupied the remaining larger region. We also found that slices containing the medial region of the SCN generated shorter circadian periods than slices that contained the lateral region of the SCN. Interestingly, the SPR corresponded well with the region where the SCN phase wave is generated. We numerically simulated the relationship between the SPR and a large LPR. A mathematical model of the SCN based on our findings faithfully reproduced the kinetics of the oscillators in the SCN in synchronized conditions, assuming the existence of clustered short-period oscillators.


Subject(s)
Circadian Clocks , Neurons/metabolism , Period Circadian Proteins/metabolism , Suprachiasmatic Nucleus/physiology , Animals , Circadian Clocks/drug effects , Colforsin/pharmacology , Male , Period Circadian Proteins/genetics , Rats , Rats, Transgenic , Rats, Wistar , Suprachiasmatic Nucleus/drug effects
16.
Genes Cells ; 18(7): 575-88, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23758111

ABSTRACT

Organisms have seasonal physiological changes in response to day length. Long-day stimulation induces thyroid-stimulating hormone beta subunit (TSHß) in the pars tuberalis (PT), which mediates photoperiodic reactions like day-length measurement and physiological adaptation. However, the mechanism of TSHß induction for day-length measurement is largely unknown. To screen candidate upstream molecules of TSHß, which convey light information to the PT, we generated Luciferase knock-in mice, which quantitatively report the dynamics of TSHß expression. We cultured brain slices containing the PT region from adult and neonatal mice and measured the bioluminescence activities from each slice over several days. A decrease in the bioluminescence activities was observed after melatonin treatment in adult and neonatal slices. These observations indicate that the experimental system possesses responsiveness of the TSHß expression to melatonin. Thus, we concluded that our experimental system monitors TSHß expression dynamics in response to external stimuli.


Subject(s)
Photoperiod , Thyrotropin, beta Subunit/metabolism , Animals , Melatonin/metabolism , Mice , Thyrotropin, beta Subunit/genetics , Time Factors
17.
J Clin Endocrinol Metab ; 98(3): E547-57, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23386640

ABSTRACT

CONTEXT: Loss-of-function mutations in PROK2 and PROKR2 have been implicated in Kallmann syndrome (KS), characterized by hypogonadotropic hypogonadism and anosmia. Recent data suggest overlapping phenotypes/genotypes between KS and congenital hypopituitarism (CH), including septo-optic dysplasia (SOD). OBJECTIVE: We screened a cohort of patients with complex forms of CH (n = 422) for mutations in PROK2 and PROKR2. RESULTS: We detected 5 PROKR2 variants in 11 patients with SOD/CH: novel p.G371R and previously reported p.A51T, p.R85L, p.L173R, and p.R268C-the latter 3 being known functionally deleterious variants. Surprisingly, 1 patient with SOD was heterozygous for the p.L173R variant, whereas his phenotypically unaffected mother was homozygous for the variant. We sought to clarify the role of PROKR2 in hypothalamopituitary development through analysis of Prokr2(-/-) mice. Interestingly, these revealed predominantly normal hypothalamopituitary development and terminal cell differentiation, with the exception of reduced LH; this was inconsistent with patient phenotypes and more analogous to the healthy mother, although she did not have KS, unlike the Prokr2(-/-) mice. CONCLUSIONS: The role of PROKR2 in the etiology of CH, SOD, and KS is uncertain, as demonstrated by no clear phenotype-genotype correlation; loss-of-function variants in heterozygosity or homozygosity can be associated with these disorders. However, we report a phenotypically normal parent, homozygous for p.L173R. Our data suggest that the variants identified herein are unlikely to be implicated in isolation in these disorders; other genetic or environmental modifiers may also impact on the etiology. Given the phenotypic variability, genetic counseling may presently be inappropriate.


Subject(s)
Gastrointestinal Hormones/genetics , Hypopituitarism/genetics , Kallmann Syndrome/genetics , Neuropeptides/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, Peptide/genetics , Septo-Optic Dysplasia/genetics , Animals , Child , Child, Preschool , Cohort Studies , Female , Genetic Testing , Genotype , HEK293 Cells , Heterozygote , Homozygote , Humans , Hypopituitarism/congenital , Hypothalamo-Hypophyseal System/embryology , Hypothalamo-Hypophyseal System/physiology , Infant , Infant, Newborn , Male , Mice , Mice, Knockout , Pedigree , Phenotype
18.
Neurosci Lett ; 535: 12-7, 2013 Feb 22.
Article in English | MEDLINE | ID: mdl-23298785

ABSTRACT

Polysialic acids are implicated in various biological processes such as neural cell migration, axonal growth, synaptogenesis and resetting of the circadian rhythm. Recently, polysialation has been reported to be involved in the formation and resetting of the circadian clock. However, the genes that control the circadian rhythm of polysialation have not been elucidated. In the present study, we investigated the expression profile of ST8 alpha-N-acetyl-neuraminide alpha-2,8-sialyltransferase 6 (ST8Sia VI) in the suprachiasmatic nucleus (SCN), which is one of the modification transferases that add sialic acids to type O carbohydrate chains. ST8Sia VI mRNA showed strong expression in the SCN with dynamic circadian rhythm. Further, the amount of ST8Sia VI mRNA in the SCN was increased by brief light exposure. Interestingly, the localization of ST8Sia VI mRNA in the SCN differs from those of arginine vasopressin and vasoactive intestinal peptide mRNAs, which are typical SCN subregion markers showing shell and core, dorsomedial and ventrolateral, or light-responsive and unresponsive regions, respectively. The present findings suggest that ST8siVI is involved in rhythmic polysialation in the SCN and that ST8siVI expression provides a novel compartmentation of the mammalian circadian center.


Subject(s)
Circadian Rhythm , Sialyltransferases/metabolism , Suprachiasmatic Nucleus/enzymology , Animals , Light , Male , Mice , Mice, Inbred BALB C , Neurons/enzymology , RNA, Messenger/metabolism , Sialyltransferases/genetics , Suprachiasmatic Nucleus/radiation effects
19.
PLoS One ; 7(9): e45373, 2012.
Article in English | MEDLINE | ID: mdl-23028969

ABSTRACT

Increased information on the encoded mammalian genome is expected to facilitate an integrated understanding of complex anatomical structure and function based on the knowledge of gene products. Determination of gene expression-anatomy associations is crucial for this understanding. To elicit the association in the three-dimensional (3D) space, we introduce a novel technique for comprehensive mapping of endogenous gene expression into a web-accessible standard space: Transcriptome Tomography. The technique is based on conjugation of sequential tissue-block sectioning, all fractions of which are used for molecular measurements of gene expression densities, and the block- face imaging, which are used for 3D reconstruction of the fractions. To generate a 3D map, tissues are serially sectioned in each of three orthogonal planes and the expression density data are mapped using a tomographic technique. This rapid and unbiased mapping technique using a relatively small number of original data points allows researchers to create their own expression maps in the broad anatomical context of the space. In the first instance we generated a dataset of 36,000 maps, reconstructed from data of 61 fractions measured with microarray, covering the whole mouse brain (ViBrism: http://vibrism.riken.jp/3dviewer/ex/index.html) in one month. After computational estimation of the mapping accuracy we validated the dataset against existing data with respect to the expression location and density. To demonstrate the relevance of the framework, we showed disease related expression of Huntington's disease gene and Bdnf. Our tomographic approach is applicable to analysis of any biological molecules derived from frozen tissues, organs and whole embryos, and the maps are spatially isotropic and well suited to the analysis in the standard space (e.g. Waxholm Space for brain-atlas databases). This will facilitate research creating and using open-standards for a molecular-based understanding of complex structures; and will contribute to new insights into a broad range of biological and medical questions.


Subject(s)
Brain/metabolism , Transcriptome/genetics , Animals , Gene Expression Profiling , Huntington Disease , Imaging, Three-Dimensional , Male , Mice , Mice, Inbred C57BL
20.
PLoS One ; 6(8): e23228, 2011.
Article in English | MEDLINE | ID: mdl-21858037

ABSTRACT

The adult mammalian brain is composed of distinct regions with specialized roles including regulation of circadian clocks, feeding, sleep/awake, and seasonal rhythms. To find quantitative differences of expression among such various brain regions, we conducted the BrainStars (B*) project, in which we profiled the genome-wide expression of ∼50 small brain regions, including sensory centers, and centers for motion, time, memory, fear, and feeding. To avoid confounds from temporal differences in gene expression, we sampled each region every 4 hours for 24 hours, and pooled the samples for DNA-microarray assays. Therefore, we focused on spatial differences in gene expression. We used informatics to identify candidate genes with expression changes showing high or low expression in specific regions. We also identified candidate genes with stable expression across brain regions that can be used as new internal control genes, and ligand-receptor interactions of neurohormones and neurotransmitters. Through these analyses, we found 8,159 multi-state genes, 2,212 regional marker gene candidates for 44 small brain regions, 915 internal control gene candidates, and 23,864 inferred ligand-receptor interactions. We also found that these sets include well-known genes as well as novel candidate genes that might be related to specific functions in brain regions. We used our findings to develop an integrated database (http://brainstars.org/) for exploring genome-wide expression in the adult mouse brain, and have made this database openly accessible. These new resources will help accelerate the functional analysis of the mammalian brain and the elucidation of its regulatory network systems.


Subject(s)
Brain/metabolism , Gene Expression Profiling/methods , Oligonucleotide Array Sequence Analysis/methods , Transcriptome , Animals , Cluster Analysis , Gene Regulatory Networks , In Situ Hybridization , Male , Mice , Mice, Inbred BALB C , Models, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...