Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Commun Biol ; 5(1): 492, 2022 05 23.
Article in English | MEDLINE | ID: mdl-35606540

ABSTRACT

Hibernation involves prolonged intervals of profound metabolic suppression periodically interrupted by brief arousals to euthermy, the function of which is unknown. Annual cycles in mammals are timed by a photoperiodically-regulated thyroid-hormone-dependent mechanism in hypothalamic tanycytes, driven by thyrotropin (TSH) in the pars tuberalis (PT), which regulates local TH-converting deiodinases and triggers remodeling of neuroendocrine pathways. We demonstrate that over the course of hibernation in continuous darkness, arctic ground squirrels (Urocitellus parryii) up-regulate the retrograde TSH/Deiodinase/TH pathway, remodel hypothalamic tanycytes, and activate the reproductive axis. Forcing the premature termination of hibernation by warming animals induced hypothalamic deiodinase expression and the accumulation of secretory granules in PT thyrotrophs and pituitary gonadotrophs, but did not further activate the reproductive axis. We suggest that periodic arousals may allow for the transient activation of hypothalamic thyroid hormone signaling, cellular remodeling, and re-programming of brain circuits in preparation for the short Arctic summer.


Subject(s)
Hibernation , Animals , Hibernation/physiology , Iodide Peroxidase , Sciuridae/physiology , Thyroid Hormones , Thyrotropin
2.
Andrology ; 9(5): 1652-1661, 2021 09.
Article in English | MEDLINE | ID: mdl-33998165

ABSTRACT

BACKGROUND: Circulating prolactin concentration in rodents and humans is sexually dimorphic. Oestrogens are a well-characterised stimulator of prolactin release. Circulating prolactin fluctuates throughout the menstrual/oestrous cycle of females in response to oestrogen levels, but remains continually low in males. We have previously identified androgens as an inhibitor of prolactin release through characterisation of males of a mouse line with a conditional pituitary androgen receptor knockout (PARKO) which have an increase in circulating prolactin, but unchanged lactotroph number. OBJECTIVES: In the present study, we aimed to specify the cell type that androgens act on to repress prolactin release. MATERIALS AND METHODS: PARKO, lactotroph-specific, Pit1 lineage-specific and neural-specific conditional androgen receptor knockout male mice were investigated using prolactin ELISA, pituitary electron microscopy, immunohistochemistry and qRT-PCR. RESULTS: Lactotroph-specific, Pit1 lineage-specific and neural-specific conditional AR knockouts did not duplicate the high circulating prolactin seen in the PARKO line. Using electron microscopy to examine ultrastructure, we showed that pituitary androgen receptor knockout male mice develop lactotrophs that resemble those seen in female mice. Castrated PARKO males have significantly reduced circulating prolactin compared to intact males. When expression of selected oestrogen-regulated anterior pituitary genes was examined, there were no differences in expression level between controls and knockouts. DISCUSSION: The cell type that androgens act on to repress prolactin release is not the lactotroph, cells in the Pit1-lineage, or the dopaminergic neurons in the hypothalamus. PARKO males develop a female-specific lactotroph ultrastructure that this is likely to contribute to the increase in circulating prolactin. Castrated PARKO males have significantly reduced circulating prolactin compared to intact males, which suggests that removal of both circulating oestrogens and androgens reduces the stimulation of pituitary prolactin release. CONCLUSION: Further investigation is needed into prolactin regulation by changes in androgen-oestrogen balance, which is involved sexual dimorphism of development and diseases including hyperprolactinemia.


Subject(s)
Hyperprolactinemia/genetics , Lactotrophs , Receptors, Androgen/deficiency , Animals , Estrogens/metabolism , Male , Mice , Mice, Knockout , Pituitary Gland/metabolism , Prolactin/metabolism
3.
J Neuroendocrinol ; 32(10): e12903, 2020 10.
Article in English | MEDLINE | ID: mdl-32959418

ABSTRACT

The anterior and intermediate lobes of the pituitary are composed of endocrine cells, as well as vasculature and supporting cells, such as folliculostellate cells. Folliculostellate cells form a network with several postulated roles in the pituitary, including production of paracrine signalling molecules and cytokines, coordination of endocrine cell hormone release, phagocytosis, and structural support. Folliculostellate cells in rats are characterised by expression of S100B protein, and in humans by glial fibrillary acid protein. However, there is evidence for another network of supporting cells in the anterior pituitary that has properties of mural cells, such as vascular smooth muscle cells and pericytes. The present study aims to characterise the distribution of cells that express the mural cell marker platelet derived growth factor receptor beta (PDGFRß) in the mouse pituitary and establish whether these cells are folliculostellate. By immunohistochemical localisation, we determine that approximately 80% of PDGFRß+ cells in the mouse pituitary have a non-perivascular location and 20% are pericytes. Investigation of gene expression in a magnetic cell sorted population of PDGFRß+ cells shows that, despite a mostly non-perivascular location, this population is enriched for mural cell markers but not enriched for rat or human folliculostellate cell markers. This is confirmed by immunohistochemistry. The present study concludes that a mural cell network is present throughout the anterior pituitary of the mouse and that this population does not express well-characterised human or rat folliculostellate cell markers.


Subject(s)
Cell Communication/physiology , Pituitary Gland/cytology , Animals , Biomarkers/metabolism , Endocrine Cells/cytology , Endocrine Cells/physiology , Endothelial Cells/cytology , Endothelial Cells/physiology , Mice , Mice, Inbred C57BL , Pericytes/cytology , Pericytes/physiology , Pituitary Gland/metabolism , Pituitary Gland, Anterior/cytology , Pituitary Gland, Anterior/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , SOXB1 Transcription Factors/metabolism
4.
JCI Insight ; 52019 07 02.
Article in English | MEDLINE | ID: mdl-31265437

ABSTRACT

Hormones produced by the anterior pituitary gland regulate an array of important physiological functions, but pituitary hormone disorders are not fully understood. Herein we report that genetically-engineered mice with deletion of the hedgehog signaling receptor Patched1 by S100a4 promoter-driven Cre recombinase (S100a4-Cre;Ptch1fl/fl mutants) exhibit adult-onset hypogonadotropic hypogonadism and multiple pituitary hormone disorders. During the transition from puberty to adult, S100a4-Cre;Ptch1fl/fl mice of both sexes develop hypogonadism coupled with reduced gonadotropin levels. Their pituitary glands also display severe structural and functional abnormalities, as revealed by transmission electron microscopy and expression of key genes regulating pituitary endocrine functions. S100a4-Cre activity in the anterior pituitary gland is restricted to CD45+ cells of hematopoietic origin, including folliculo-stellate cells and other immune cell types, causing sex-specific changes in the expression of genes regulating the local microenvironment of the anterior pituitary. These findings provide in vivo evidence for the importance of pituitary hematopoietic cells in regulating fertility and endocrine function, in particular during sexual maturation and likely through sexually dimorphic mechanisms. These findings support a previously unrecognized role of hematopoietic cells in causing hypogonadotropic hypogonadism and provide inroads into the molecular and cellular basis for pituitary hormone disorders in humans.


Subject(s)
Hypogonadism/metabolism , Integrases/metabolism , Patched-1 Receptor/metabolism , Pituitary Gland/metabolism , S100 Calcium-Binding Protein A4/metabolism , Animals , Epididymis/pathology , Female , Humans , Hypogonadism/genetics , Hypogonadism/pathology , Male , Mice , Mice, Knockout , Ovary/pathology , Patched-1 Receptor/genetics , Pituitary Gland, Anterior/metabolism , Reproduction/physiology , Seminal Vesicles/pathology , Sexual Maturation , Signal Transduction , Testis , Testosterone/blood , Uterus/pathology
5.
PLoS Comput Biol ; 15(6): e1007030, 2019 06.
Article in English | MEDLINE | ID: mdl-31194728

ABSTRACT

Prolactin is a major hormone product of the pituitary gland, the central endocrine regulator. Despite its physiological importance, the cell-level mechanisms of prolactin production are not well understood. Having significantly improved the resolution of real-time-single-cell-GFP-imaging, the authors recently revealed that prolactin gene transcription is highly dynamic and stochastic yet shows space-time coordination in an intact tissue slice. However, it still remains an open question as to what kind of cellular communication mediates the observed space-time organization. To determine the type of interaction between cells we developed a statistical model. The degree of similarity between two expression time series was studied in terms of two distance measures, Euclidean and geodesic, the latter being a network-theoretic distance defined to be the minimal number of edges between nodes, and this was used to discriminate between juxtacrine from paracrine signalling. The analysis presented here suggests that juxtacrine signalling dominates. To further determine whether the coupling is coordinating transcription or post-transcriptional activities we used stochastic switch modelling to infer the transcriptional profiles of cells and estimated their similarity measures to deduce that their spatial cellular coordination involves coupling of transcription via juxtacrine signalling. We developed a computational model that involves an inter-cell juxtacrine coupling, yielding simulation results that show space-time coordination in the transcription level that is in agreement with the above analysis. The developed model is expected to serve as the prototype for the further study of tissue-level organised gene expression for epigenetically regulated genes, such as prolactin.


Subject(s)
Cell Communication/genetics , Models, Biological , Paracrine Communication/genetics , Animals , Cell Communication/physiology , Computational Biology , Gene Expression Regulation/genetics , Humans , Male , Paracrine Communication/physiology , Pituitary Gland/metabolism , Prolactin/genetics , Prolactin/metabolism , Rats , Rats, Transgenic , Stochastic Processes
6.
Cereb Cortex ; 29(5): 2148-2159, 2019 05 01.
Article in English | MEDLINE | ID: mdl-29850799

ABSTRACT

Synaptosomal associated protein 25 kDa (SNAP25) is an essential component of the SNARE complex regulating synaptic vesicle fusion. SNAP25 deficiency has been implicated in a variety of cognitive disorders. We ablated SNAP25 from selected neuronal populations by generating a transgenic mouse (B6-Snap25tm3mcw (Snap25-flox)) with LoxP sites flanking exon5a/5b. In the presence of Cre-recombinase, Snap25-flox is recombined to a truncated transcript. Evoked synaptic vesicle release is severely reduced in Snap25 conditional knockout (cKO) neurons as shown by live cell imaging of synaptic vesicle fusion and whole cell patch clamp recordings in cultured hippocampal neurons. We studied Snap25 cKO in subsets of cortical projection neurons in vivo (L5-Rbp4-Cre; L6-Ntsr1-Cre; L6b-Drd1a-Cre). cKO neurons develop normal axonal projections, but axons are not maintained appropriately, showing signs of swelling, fragmentation and eventually complete absence. Onset and progression of degeneration are dependent on the neuron type, with L5 cells showing the earliest and most severe axonal loss. Ultrastructural examination revealed that cKO neurites contain autophagosome/lysosome-like structures. Markers of inflammation such as Iba1 and lipofuscin are increased only in adult cKO cortex. Snap25 cKO can provide a model to study genetic interactions with environmental influences in several disorders.


Subject(s)
Brain/growth & development , Brain/pathology , Neurons/pathology , Neurons/physiology , Synaptosomal-Associated Protein 25/physiology , Animals , Axons/pathology , Axons/physiology , Axons/ultrastructure , Brain/ultrastructure , Female , Male , Mice, Knockout , Neurons/ultrastructure , Synaptic Transmission , Synaptic Vesicles
7.
Sci Rep ; 7: 45541, 2017 03 31.
Article in English | MEDLINE | ID: mdl-28361877

ABSTRACT

Despite being unable to activate the cognate ghrelin receptor (GHS-R), unacylated ghrelin (UAG) possesses a unique activity spectrum that includes promoting bone marrow adipogenesis. Since a receptor mediating this action has not been identified, we re-appraised the potential interaction of UAG with GHS-R in the regulation of bone marrow adiposity. Surprisingly, the adipogenic effects of intra-bone marrow (ibm)-infused acylated ghrelin (AG) and UAG were abolished in male GHS-R-null mice. Gas chromatography showed that isolated tibial marrow adipocytes contain the medium-chain fatty acids utilised in the acylation of UAG, including octanoic acid. Additionally, immunohistochemistry and immunogold electron microscopy revealed that tibial marrow adipocytes show prominent expression of the UAG-activating enzyme ghrelin O-acyl transferase (GOAT), which is located in the membranes of lipid trafficking vesicles and in the plasma membrane. Finally, the adipogenic effect of ibm-infused UAG was completely abolished in GOAT-KO mice. Thus, the adipogenic action of exogenous UAG in tibial marrow is dependent upon acylation by GOAT and activation of GHS-R. This suggests that UAG is subject to target cell-mediated activation - a novel mechanism for manipulating hormone activity.


Subject(s)
Acyltransferases/metabolism , Adipogenesis , Bone Marrow/metabolism , Ghrelin/metabolism , Membrane Proteins/metabolism , Receptors, Ghrelin/metabolism , Acylation , Animals , Chromatography, Gas , Immunohistochemistry , Mice , Mice, Knockout , Microscopy, Immunoelectron , Receptors, Ghrelin/deficiency
8.
Am J Physiol Renal Physiol ; 312(4): F778-F790, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28179256

ABSTRACT

Zebrafish provide an excellent model in which to assess the role of the renin-angiotensin system in renal development, injury, and repair. In contrast to mammals, zebrafish kidney organogenesis terminates with the mesonephros. Despite this, the basic functional structure of the nephron is conserved across vertebrates. The relevance of teleosts for studies relating to the regulation of the renin-angiotensin system was established by assessing the phenotype and functional regulation of renin-expressing cells in zebrafish. Transgenic fluorescent reporters for renin (ren), smooth muscle actin (acta2), and platelet-derived growth factor receptor-beta (pdgfrb) were studied to determine the phenotype and secretory ultrastructure of perivascular renin-expressing cells. Whole kidney ren transcription responded to altered salinity, pharmacological renin-angiotensin system inhibition, and renal injury. Mesonephric ren-expressing cells occupied niches at the preglomerular arteries and afferent arterioles, forming intermittent epithelioid-like multicellular clusters exhibiting a granular secretory ultrastructure. In contrast, renin cells of the efferent arterioles were thin bodied and lacked secretory granules. Renin cells expressed the perivascular cell markers acta2 and pdgfrb Transcriptional responses of ren to physiological challenge support the presence of a functional renin-angiotensin system and are consistent with the production of active renin. The reparative capability of the zebrafish kidney was harnessed to demonstrate that ren transcription is a marker for renal injury and repair. Our studies demonstrate substantive conservation of renin regulation across vertebrates, and ultrastructural studies of renin cells reveal at least two distinct morphologies of mesonephric perivascular ren-expressing cells.


Subject(s)
Cell Shape , Renin-Angiotensin System , Renin/metabolism , Wolffian Ducts/enzymology , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Actins/genetics , Actins/metabolism , Animals , Animals, Genetically Modified , Gene Expression Regulation, Developmental , Genotype , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Microscopy, Fluorescence , Myocytes, Smooth Muscle/metabolism , Pericytes/metabolism , Phenotype , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptor, Platelet-Derived Growth Factor beta/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Renin/genetics , Transcription, Genetic , Wolffian Ducts/ultrastructure , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics
9.
Sci Rep ; 7: 40620, 2017 01 17.
Article in English | MEDLINE | ID: mdl-28094777

ABSTRACT

Nicotinic Acid Adenine Dinucleotide Phosphate (NAADP) stimulates calcium release from acidic stores such as lysosomes and is a highly potent calcium-mobilising second messenger. NAADP plays an important role in calcium signalling in the heart under basal conditions and following ß-adrenergic stress. Nevertheless, the spatial interaction of acidic stores with other parts of the calcium signalling apparatus in cardiac myocytes is unknown. We present evidence that lysosomes are intimately associated with the sarcoplasmic reticulum (SR) in ventricular myocytes; a median separation of 20 nm in 2D electron microscopy and 3.3 nm in 3D electron tomography indicates a genuine signalling microdomain between these organelles. Fourier analysis of immunolabelled lysosomes suggests a sarcomeric pattern (dominant wavelength 1.80 µm). Furthermore, we show that lysosomes form close associations with mitochondria (median separation 6.2 nm in 3D studies) which may provide a basis for the recently-discovered role of NAADP in reperfusion-induced cell death. The trigger hypothesis for NAADP action proposes that calcium release from acidic stores subsequently acts to enhance calcium release from the SR. This work provides structural evidence in cardiac myocytes to indicate the formation of microdomains between acidic and SR calcium stores, supporting emerging interpretations of NAADP physiology and pharmacology in heart.


Subject(s)
Lysosomes/metabolism , Lysosomes/ultrastructure , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/ultrastructure , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum/ultrastructure , Animals , Biomarkers , Calcium/metabolism , Calcium Channels/metabolism , Calcium Signaling , Heart Ventricles/cytology , Heart Ventricles/metabolism , Lysosomal-Associated Membrane Protein 2/metabolism , Male , NADP/analogs & derivatives , NADP/metabolism , Organelles/metabolism , Rabbits
10.
Elife ; 52016 11 29.
Article in English | MEDLINE | ID: mdl-27897970

ABSTRACT

Hepcidin is the master regulator of systemic iron homeostasis. Derived primarily from the liver, it inhibits the iron exporter ferroportin in the gut and spleen, the sites of iron absorption and recycling respectively. Recently, we demonstrated that ferroportin is also found in cardiomyocytes, and that its cardiac-specific deletion leads to fatal cardiac iron overload. Hepcidin is also expressed in cardiomyocytes, where its function remains unknown. To define the function of cardiomyocyte hepcidin, we generated mice with cardiomyocyte-specific deletion of hepcidin, or knock-in of hepcidin-resistant ferroportin. We find that while both models maintain normal systemic iron homeostasis, they nonetheless develop fatal contractile and metabolic dysfunction as a consequence of cardiomyocyte iron deficiency. These findings are the first demonstration of a cell-autonomous role for hepcidin in iron homeostasis. They raise the possibility that such function may also be important in other tissues that express both hepcidin and ferroportin, such as the kidney and the brain.


Subject(s)
Hepcidins/metabolism , Homeostasis , Iron/metabolism , Myocytes, Cardiac/metabolism , Animals , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Gene Knock-In Techniques , Gene Knockout Techniques , Hepcidins/genetics , Mice
11.
Mol Cell ; 64(4): 746-759, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27863227

ABSTRACT

Excitation-transcription coupling, linking stimulation at the cell surface to changes in nuclear gene expression, is conserved throughout eukaryotes. How closely related coexpressed transcription factors are differentially activated remains unclear. Here, we show that two Ca2+-dependent transcription factor isoforms, NFAT1 and NFAT4, require distinct sub-cellular InsP3 and Ca2+ signals for physiologically sustained activation. NFAT1 is stimulated by sub-plasmalemmal Ca2+ microdomains, whereas NFAT4 additionally requires Ca2+ mobilization from the inner nuclear envelope by nuclear InsP3 receptors. NFAT1 is rephosphorylated (deactivated) more slowly than NFAT4 in both cytoplasm and nucleus, enabling a more prolonged activation phase. Oscillations in cytoplasmic Ca2+, long considered the physiological form of Ca2+ signaling, play no role in activating either NFAT protein. Instead, effective sustained physiological activation of NFAT4 is tightly linked to oscillations in nuclear Ca2+. Our results show how gene expression can be controlled by coincident yet geographically distinct Ca2+ signals, generated by a freely diffusible InsP3 message.


Subject(s)
Calcium Signaling , Calcium/metabolism , Inositol Phosphates/metabolism , NFATC Transcription Factors/genetics , Recombinant Fusion Proteins/genetics , Animals , Basophils/cytology , Basophils/drug effects , Basophils/metabolism , Bronchi/cytology , Bronchi/drug effects , Bronchi/metabolism , Cell Line , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Inositol 1,4,5-Trisphosphate Receptors/genetics , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Leukotriene C4/pharmacology , NFATC Transcription Factors/metabolism , Protein Transport , Rats , Recombinant Fusion Proteins/metabolism , Thapsigargin/pharmacology , Transcription, Genetic
12.
Kidney Int ; 90(6): 1251-1261, 2016 12.
Article in English | MEDLINE | ID: mdl-27678158

ABSTRACT

Pericytes, perivascular cells embedded in the microvascular wall, are crucial for vascular homeostasis. These cells also play diverse roles in tissue development and regeneration as multi-lineage progenitors, immunomodulatory cells and as sources of trophic factors. Here, we establish that pericytes are renin producing cells in the human kidney. Renin was localized by immunohistochemistry in CD146 and NG2 expressing pericytes, surrounding juxtaglomerular and afferent arterioles. Similar to pericytes from other organs, CD146+CD34-CD45-CD56- renal fetal pericytes, sorted by flow cytometry, exhibited tri-lineage mesodermal differentiation potential in vitro. Additionally, renin expression was triggered in cultured kidney pericytes by cyclic AMP as confirmed by immuno-electron microscopy, and secretion of enzymatically functional renin, capable of generating angiotensin I. Pericytes derived from second trimester human placenta also expressed renin in an inducible fashion although the renin activity was much lower than in renal pericytes. Thus, our results confirm and extend the recently discovered developmental plasticity of microvascular pericytes, and may open new perspectives to the therapeutic regulation of the renin-angiotensin system.


Subject(s)
Kidney/ultrastructure , Pericytes/metabolism , Renin/metabolism , Humans , Kidney/embryology , Mesenchymal Stem Cells , Primary Cell Culture
13.
Endocrinology ; 157(9): 3631-46, 2016 09.
Article in English | MEDLINE | ID: mdl-27580811

ABSTRACT

Thyrotrope hyperplasia and hypertrophy are common responses to primary hypothyroidism. To understand the genetic regulation of these processes, we studied gene expression changes in the pituitaries of Cga(-/-) mice, which are deficient in the common α-subunit of TSH, LH, and FSH. These mice have thyrotrope hypertrophy and hyperplasia and develop thyrotrope adenoma. We report that cell proliferation is increased, but the expression of most stem cell markers is unchanged. The α-subunit is required for secretion of the glycoprotein hormone ß-subunits, and mutants exhibit elevated expression of many genes involved in the unfolded protein response, consistent with dilation and stress of the endoplasmic reticulum. Mutants have elevated expression of transcription factors that are important in thyrotrope function, such as Gata2 and Islet 1, and those that stimulate proliferation, including Nupr1, E2f1, and Etv5. We characterized the expression and function of a novel, overexpressed gene, transcription elongation factor A (SII)-like 5 (Tceal5). Stable expression of Tceal5 in a pituitary progenitor cell line is sufficient to increase cell proliferation. Thus, Tceal5 may act as a proto-oncogene. This study provides a rich resource for comparing pituitary transcriptomes and an analysis of gene expression networks.


Subject(s)
Adenoma/metabolism , Hypothyroidism/metabolism , Pituitary Neoplasms/metabolism , Thyrotrophs/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Cell Proliferation , Endoplasmic Reticulum/ultrastructure , Endoplasmic Reticulum Stress , Female , Gene Expression , Gene Expression Profiling , Glycoprotein Hormones, alpha Subunit/genetics , Male , Mice , Molecular Sequence Data , Proto-Oncogene Mas , Thyrotropin, beta Subunit/metabolism , Transcriptional Elongation Factors/metabolism , Unfolded Protein Response
14.
Stem Cell Reports ; 6(3): 342-56, 2016 Mar 08.
Article in English | MEDLINE | ID: mdl-26905200

ABSTRACT

Heterozygous mutations in the glucocerebrosidase gene (GBA) represent the strongest common genetic risk factor for Parkinson's disease (PD), the second most common neurodegenerative disorder. However, the molecular mechanisms underlying this association are still poorly understood. Here, we have analyzed ten independent induced pluripotent stem cell (iPSC) lines from three controls and three unrelated PD patients heterozygous for the GBA-N370S mutation, and identified relevant disease mechanisms. After differentiation into dopaminergic neurons, we observed misprocessing of mutant glucocerebrosidase protein in the ER, associated with activation of ER stress and abnormal cellular lipid profiles. Furthermore, we observed autophagic perturbations and an enlargement of the lysosomal compartment specifically in dopamine neurons. Finally, we found increased extracellular α-synuclein in patient-derived neuronal culture medium, which was not associated with exosomes. Overall, ER stress, autophagic/lysosomal perturbations, and elevated extracellular α-synuclein likely represent critical early cellular phenotypes of PD, which might offer multiple therapeutic targets.


Subject(s)
Autophagy , Dopaminergic Neurons/metabolism , Endoplasmic Reticulum Stress , Glucosylceramidase/genetics , Induced Pluripotent Stem Cells/cytology , Parkinson Disease/metabolism , alpha-Synuclein/metabolism , Animals , Cell Line , Cells, Cultured , Dopaminergic Neurons/cytology , Exosomes/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Lysosomes/metabolism , Mice , Mutation, Missense , Neurogenesis , Parkinson Disease/genetics , Parkinson Disease/pathology
15.
Elife ; 5: e08494, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26828110

ABSTRACT

Transcription at individual genes in single cells is often pulsatile and stochastic. A key question emerges regarding how this behaviour contributes to tissue phenotype, but it has been a challenge to quantitatively analyse this in living cells over time, as opposed to studying snap-shots of gene expression state. We have used imaging of reporter gene expression to track transcription in living pituitary tissue. We integrated live-cell imaging data with statistical modelling for quantitative real-time estimation of the timing of switching between transcriptional states across a whole tissue. Multiple levels of transcription rate were identified, indicating that gene expression is not a simple binary 'on-off' process. Immature tissue displayed shorter durations of high-expressing states than the adult. In adult pituitary tissue, direct cell contacts involving gap junctions allowed local spatial coordination of prolactin gene expression. Our findings identify how heterogeneous transcriptional dynamics of single cells may contribute to overall tissue behaviour.


Subject(s)
Gene Expression Regulation , Pituitary Gland/physiology , Transcription, Genetic , Animals , Gene Expression Profiling , Genes, Reporter , Optical Imaging , Rats, Inbred F344 , Spatio-Temporal Analysis
16.
Curr Biol ; 25(20): 2651-62, 2015 Oct 19.
Article in English | MEDLINE | ID: mdl-26412130

ABSTRACT

Persistent free-running circannual (approximately year-long) rhythms have evolved in animals to regulate hormone cycles, drive metabolic rhythms (including hibernation), and time annual reproduction. Recent studies have defined the photoperiodic input to this rhythm, wherein melatonin acts on thyrotroph cells of the pituitary pars tuberalis (PT), leading to seasonal changes in the control of thyroid hormone metabolism in the hypothalamus. However, seasonal rhythms persist in constant conditions in many species in the absence of a changing photoperiod signal, leading to the generation of circannual cycles. It is not known which cells, tissues, and pathways generate these remarkable long-term rhythmic processes. We show that individual PT thyrotrophs can be in one of two binary states reflecting either a long (EYA3(+)) or short (CHGA(+)) photoperiod, with the relative proportion in each state defining the phase of the circannual cycle. We also show that a morphogenic cycle driven by the PT leads to extensive re-modeling of the PT and hypothalamus over the circannual cycle. We propose that the PT may employ a recapitulated developmental pathway to drive changes in morphology of tissues and cells. Our data are consistent with the hypothesis that the circannual timer may reside within the PT thyrotroph and is encoded by a binary switch timing mechanism, which may regulate the generation of circannual neuroendocrine rhythms, leading to dynamic re-modeling of the hypothalamic interface. In summary, the PT-ventral hypothalamus now appears to be a prime structure involved in long-term rhythm generation.


Subject(s)
Circadian Clocks , Photoperiod , Sheep/physiology , Thyrotrophs/physiology , Animals , Male
17.
Proc Natl Acad Sci U S A ; 112(10): 3164-9, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25713362

ABSTRACT

Iron is essential to the cell. Both iron deficiency and overload impinge negatively on cardiac health. Thus, effective iron homeostasis is important for cardiac function. Ferroportin (FPN), the only known mammalian iron-exporting protein, plays an essential role in iron homeostasis at the systemic level. It increases systemic iron availability by releasing iron from the cells of the duodenum, spleen, and liver, the sites of iron absorption, recycling, and storage respectively. However, FPN is also found in tissues with no known role in systemic iron handling, such as the heart, where its function remains unknown. To explore this function, we generated mice with a cardiomyocyte-specific deletion of Fpn. We show that these animals have severely impaired cardiac function, with a median survival of 22 wk, despite otherwise unaltered systemic iron status. We then compared their phenotype with that of ubiquitous hepcidin knockouts, a recognized model of the iron-loading disease hemochromatosis. The phenotype of the hepcidin knockouts was far milder, with normal survival up to 12 mo, despite far greater iron loading in the hearts. Histological examination demonstrated that, although cardiac iron accumulates within the cardiomyocytes of Fpn knockouts, it accumulates predominantly in other cell types in the hepcidin knockouts. We conclude, first, that cardiomyocyte FPN is essential for intracellular iron homeostasis and, second, that the site of deposition of iron within the heart determines the severity with which it affects cardiac function. Both findings have significant implications for the assessment and treatment of cardiac complications of iron dysregulation.


Subject(s)
Cation Transport Proteins/physiology , Heart/physiology , Homeostasis , Iron/metabolism , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout
18.
Dev Biol ; 401(2): 287-98, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25557622

ABSTRACT

Maintaining follicle integrity during development, whereby each follicle is a functional unit containing a single oocyte, is essential for the generation of healthy oocytes. However, the mechanisms that regulate this critical function have not been determined. In this paper we investigate the role of the oocyte in maintaining follicle development. To investigate this role, we use a mouse model with oocyte-specific deletion of C1galt1 which is required for the generation of core 1-derived O-glycans. The loss of oocyte-generated O-glycans results in the joining of follicles and the generation of Multiple-Oocyte Follicles (MOFs). The aim was to determine how Mutant follicle development is modified thus enabling follicles to join. Extracellular matrix and follicle permeability were studied using histology, immunohistochemistry and electron microscopy (EM). In ovaries containing Mutant Oocytes, the Follicle basal lamina (FBL) is altered both functionally and structurally from the primary stage onwards with Mutant follicles possessing unexpectedly thicker FBL. In Mutant ovaries, the theca cell layer is also modified with intermingling of theca between adjacent follicles. MOF function was analysed but despite increased numbers of preantral MOFs in Mutants, these do not reach the preovulatory stage after gonadotrophin stimulation. We propose a model describing how oocyte initiated changes in FBL and theca cells result in follicles joining. These data reveal new and important roles for the oocyte in follicle development and follicle integrity.


Subject(s)
Basement Membrane/embryology , Galactosyltransferases/genetics , Oocytes/metabolism , Ovarian Follicle/embryology , Theca Cells/cytology , Animals , Basement Membrane/cytology , Collagen/metabolism , Extracellular Matrix/metabolism , Female , Glycoproteins/metabolism , Gonadotropins/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Ovarian Follicle/cytology , Permeability , Polysaccharides/genetics
19.
Endocrinology ; 156(3): 975-86, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25545384

ABSTRACT

Intracellular calcium-permeable channels have been implicated in thermogenic function of murine brown and brite/beige adipocytes, respectively transient receptor potential melastin-8 and transient receptor potential vanilloid-4. Because the endo-lysosomal two-pore channels (TPCs) have also been ascribed with metabolic functionality, we studied the effect of simultaneously knocking out TPC1 and TPC2 on body composition and energy balance in male mice fed a chow diet. Compared with wild-type mice, TPC1 and TPC2 double knockout (Tpcn1/2(-/-)) animals had a higher respiratory quotient and became obese between 6 and 9 months of age. Although food intake was unaltered, interscapular brown adipose tissue (BAT) maximal temperature and lean-mass adjusted oxygen consumption were lower in Tpcn1/2(-/-) than in wild type mice. Phosphorylated hormone-sensitive lipase expression, lipid density and expression of ß-adrenergic receptors were also lower in Tpcn1/2(-/-) BAT, whereas mitochondrial respiratory chain function and uncoupling protein-1 expression remained intact. We conclude that Tpcn1/2(-/-) mice show mature-onset obesity due to reduced lipid availability and use, and a defect in ß-adrenergic receptor signaling, leading to impaired thermogenic activity, in BAT.


Subject(s)
Adipose Tissue, Brown/physiology , Body Temperature Regulation/physiology , Calcium Channels/metabolism , Lipid Metabolism/physiology , Obesity/genetics , Animals , Calcium Channels/genetics , Gene Expression Regulation/physiology , Lipid Metabolism/genetics , Male , Mice , Mice, Knockout , Obesity/metabolism , Protozoan Proteins , Receptors, Adrenergic, beta/physiology , Signal Transduction
20.
J Appl Physiol (1985) ; 116(7): 885-92, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24030664

ABSTRACT

The hypoxia-inducible factor (HIF) family of transcription factors coordinates diverse cellular and systemic responses to hypoxia. Chuvash polycythemia (CP) is an autosomal recessive disorder in humans in which there is impaired oxygen-dependent degradation of HIF, resulting in long-term systemic elevation of HIF levels at normal oxygen tensions. CP patients demonstrate the characteristic features of ventilatory acclimatization to hypoxia, namely, an elevated baseline ventilation and enhanced acute hypoxic ventilatory response (AHVR). We investigated the ventilatory and carotid-body phenotype of a mouse model of CP, using whole-body plethysmography, immunohistochemistry, and electron microscopy. In keeping with studies in humans, CP mice had elevated ventilation in euoxia and a significantly exaggerated AHVR when exposed to 10% oxygen, with or without the addition of 3% carbon dioxide. Carotid-body immunohistochemistry demonstrated marked hyperplasia of the oxygen-sensing type I cells, and the cells themselves appeared enlarged with more prominent nuclei. This hypertrophy was confirmed by electron microscopy, which also revealed that the type I cells contained an increased number of mitochondria, enlarged dense-cored vesicles, and markedly expanded rough endoplasmic reticulum. The morphological and ultrastructural changes seen in the CP mouse carotid body are strikingly similar to those observed in animals exposed to chronic hypoxia. Our study demonstrates that the HIF pathway plays a major role, not only in regulating both euoxic ventilatory control and the sensitivity of the response to hypoxia, but also in determining the morphology of the carotid body.


Subject(s)
Carotid Body/pathology , Hypoxia/genetics , Lung/physiopathology , Mutation , Pulmonary Ventilation , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Acclimatization , Altitude , Animals , Carotid Body/metabolism , Carotid Body/physiopathology , Disease Models, Animal , Genotype , Hyperplasia , Hypertrophy , Hypoxia/metabolism , Hypoxia/pathology , Hypoxia/physiopathology , Lung/metabolism , Male , Mice , Mice, Mutant Strains , Phenotype , Time Factors , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...