Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Fluids Barriers CNS ; 19(1): 20, 2022 Mar 05.
Article in English | MEDLINE | ID: mdl-35248089

ABSTRACT

BACKGROUND: Hydrocephalus (increased ventricular size due to CSF accumulation) is a common finding in human ciliopathies and in mouse models with genetic depletion of the multiciliated cell (MCC) cilia machinery. However, the contribution of MCC to CSF dynamics and, the mechanism by which impaired MCC function leads to hydrocephalus remains poorly understood. The aim of our study was to examine if defects in MCC ciliogenesis and cilia-generated CSF flow impact central nervous system (CNS) fluid homeostasis including glymphatic transport and solute waste drainage. METHODS: We used two distinct mouse models of MCC ciliopathy: MCC-specific CEP164 conditional knockout mice (FOXJ1-Cre;CEP164fl/fl (N = 10), 3-month-old) and p73 knock-out (p73-/- (N = 8), 5-month-old) mice. Age-matched, wild-type littermates for each of the mutants served as controls. Glymphatic transport and solute drainage was quantified using in vivo T1 mapping by magnetic resonance imaging (MRI) after CSF infusion of gadoteric acid. Brain morphometry and aquaporin 4 expression (AQP4) was also assessed. Intracranial pressure (ICP) was measured in separate cohorts. RESULTS: In both of the two models of MCC ciliopathy we found the ventriculomegaly to be associated with normal ICP. We showed that FOXJ1-Cre;CEP164fl/fl mice with hydrocephalus still demonstrated sustained glymphatic transport and normal AQP4 expression along capillaries. In p73-/- mice glymphatic transport was even increased, and this was paralleled by an increase in AQP4 polarization around capillaries. Further, solute drainage via the cribriform plate to the nasal cavity was severely impaired in both ciliopathy models and associated with chronic rhinitis and olfactory bulb hypoplasia. CONCLUSIONS: The combination of sustained glymphatic transport, impaired solute drainage via the cribriform plate to the nasal cavity and hydrocephalus has not previously been reported in models of MCC ciliopathy. Our data enhance our understanding of how different types of ciliopathies contribute to disruption of CNS fluid homeostasis, manifested in pathologies such as hydrocephalus.


Subject(s)
Ciliopathies , Glymphatic System , Hydrocephalus , Animals , Ciliopathies/genetics , Ciliopathies/pathology , Drainage , Glymphatic System/physiology , Hydrocephalus/pathology , Mice , Nasal Cavity/pathology
2.
Life Sci Alliance ; 4(5)2021 05.
Article in English | MEDLINE | ID: mdl-33653689

ABSTRACT

Clearance of the airway is dependent on directional mucus flow across the mucociliary epithelium, and deficient flow is implicated in a range of human disorders. Efficient flow relies on proper polarization of the multiciliated cells and sufficient ciliary beat frequency. We show that NO, produced by nNOS in the multiciliated cells of the mouse trachea, controls both the planar polarity and the ciliary beat frequency and is thereby necessary for the generation of the robust flow. The effect of nNOS on the polarity of ciliated cells relies on its interactions with the apical networks of actin and microtubules and involves RhoA activation. The action of nNOS on the beat frequency is mediated by guanylate cyclase; both NO donors and cGMP can augment fluid flow in the trachea and rescue the deficient flow in nNOS mutants. Our results link insufficient availability of NO in ciliated cells to defects in flow and ciliary activity and may thereby explain the low levels of exhaled NO in ciliopathies.


Subject(s)
Cilia/metabolism , Nitric Oxide Synthase Type I/metabolism , Trachea/metabolism , Animals , Cell Polarity , Cilia/physiology , Epithelial Cells , Female , Male , Mice , Mice, Knockout , Mucus , Nitric Oxide Synthase Type I/physiology , Trachea/cytology , Trachea/physiology
3.
PLoS One ; 13(6): e0194585, 2018.
Article in English | MEDLINE | ID: mdl-29874225

ABSTRACT

Vascular smooth muscle cells (SMCs), distinguished by the expression of the neuronal stem cell marker nestin, may represent stem cell-like progenitor cells in various organs including the testis. We investigated epididymal tissues of adult nestin-GFP mice, rats after Leydig cell depletion via ethane dimethane sulfonate (EDS), rats and mice during postnatal development and human tissues. By use of Clarity, a histochemical method to illustrate a three-dimensional picture, we could demonstrate nestin-GFP positive cells within the vascular network. We localized nestin in the epididymis in proliferating vascular SMCs by colocalization with both smooth muscle actin and PCNA, and it was distinct from CD31-positive endothelial cells. The same nestin localization was found in the human epididymis. However, nestin was not found in SMCs of the epididymal duct. Nestin expression is high during postnatal development of mouse and rat and down-regulated towards adulthood when testosterone levels increase. Nestin increases dramatically in rats after Leydig cell ablation with EDS and subsequently low testosterone levels. Interestingly, during this period, the expression of androgen receptor in the epididymis is low and increases until nestin reaches normal levels of adulthood. Here we show that nestin, a common marker for neuronal stem cells, is also expressed in the vasculature of the epididymis. Our results give new insights into the yet underestimated role of proliferating nestin-expressing vascular SMCs during postnatal development and repair of the epididymis.


Subject(s)
Gene Expression Regulation , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Nestin/biosynthesis , Testosterone/deficiency , Animals , Epididymis/blood supply , Epididymis/growth & development , Epididymis/pathology , Male , Mice , Mice, Transgenic , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology
4.
J Immunol ; 197(7): 2686-94, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27574301

ABSTRACT

During embryogenesis, lymph nodes form through intimate interaction between lymphoid tissue inducer and lymphoid tissue organizer (LTo) cells. Shortly after birth in mice, specialized stromal cell subsets arise that organize microenvironments within the lymph nodes; however, their direct precursors have not yet been identified. In the bone marrow, mesenchymal stem cells are labeled with GFP in nestin-GFP mice, and we show that during all stages of development, nestin(+) cells are present within lymph nodes of these mice. At day of birth, both mesenchymal CD31(-) and endothelial CD31(+) LTo cells were GFP(+), and only the population of CD31(-) LTo cells contained mesenchymal precursors. These CD31(-)nestin(+) cells are found in the T and B cell zones or in close association with high endothelial venules in adult lymph nodes. Fate mapping of nestin(+) cells unambiguously revealed the contribution of nestin(+) precursor cells to the mesenchymal as well as the endothelial stromal populations within lymph nodes. However, postnatal tamoxifen induced targeting of nestin(+) cells in nes-creER mice showed that most endothelial cells and only a minority of the nonendothelial cells were labeled. Overall our data show that nestin(+) cells contribute to all subsets of the complex stromal populations that can be found in lymph nodes.


Subject(s)
Endothelial Cells/cytology , Lymph Nodes/cytology , Nestin/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Stromal Cells/cytology , Stromal Cells/metabolism , Animals , Gene Expression Profiling , Mice , Mice, Inbred C57BL , Nestin/genetics
6.
Eur Respir J ; 47(3): 876-88, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26699726

ABSTRACT

Nestin, a well-known marker of neuronal stem cells, was recently suggested to characterise stem cell-like progenitors in non-neuronal structures during development and tissue repair. Integrating novel morphological approaches (CLARITY), we investigate whether nestin expression defines the proliferating cell population that essentially drives vascular remodelling during development of pulmonary hypertension.The role of nestin was investigated in lungs of nestin-GFP (green fluorescent protein) mice, models of pulmonary hypertension (rat: monocrotaline, SU5416/hypoxia; mouse: hypoxia), samples from pulmonary hypertension patients and human pulmonary vascular smooth muscle cells (VSMCs).Nestin was solely found in lung vasculature and localised to proliferating VSMCs, but not bronchial smooth muscle cells. Nestin was shown to affect cell number and was significantly enhanced in lungs early during development of pulmonary hypertension, correlating well with increased VSMC proliferation, expression of phosphorylated (activated) platelet-derived growth factor receptor ß and downregulation of the smooth muscle cell differentiation marker calponin. At later time points when pulmonary hypertension became clinically evident, nestin expression and proliferation returned to control levels. Increase of nestin-positive VSMCs was also found in human pulmonary hypertension, both in vessel media and neointima.Nestin expression seems to be obligatory for VSMC proliferation, and specifies lung vascular wall cells that drive remodelling and (re-)generation. Our data promise novel diagnostic tools and therapeutic targets for pulmonary hypertension.


Subject(s)
Cell Differentiation , Cell Proliferation , Hypertension, Pulmonary/metabolism , Muscle, Smooth, Vascular/metabolism , Nestin/metabolism , Vascular Remodeling , Animals , Calcium-Binding Proteins/metabolism , Cells, Cultured , Green Fluorescent Proteins/analysis , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/metabolism , Monocrotaline , Rats , Rats, Sprague-Dawley , Receptor, Platelet-Derived Growth Factor beta/metabolism , Calponins
7.
Front Neurosci ; 9: 443, 2015.
Article in English | MEDLINE | ID: mdl-26648838

ABSTRACT

Repeated experience of winning in a social conflict setting elevates levels of aggression and may lead to violent behavioral patterns. Here, we use a paradigm of repeated aggression and fighting deprivation to examine changes in behavior, neurogenesis, and neuronal activity in mice with positive fighting experience. We show that for males, repeated positive fighting experience induces persistent demonstration of aggression and stereotypic behaviors in daily agonistic interactions, enhances aggressive motivation, and elevates levels of anxiety. When winning males are deprived of opportunities to engage in further fights, they demonstrate increased levels of aggressiveness. Positive fighting experience results in increased levels of progenitor cell proliferation and production of young neurons in the hippocampus. This increase is not diminished after a fighting deprivation period. Furthermore, repeated winning experience decreases the number of activated (c-fos-positive) cells in the basolateral amygdala and increases the number of activated cells in the hippocampus; a subsequent no-fight period restores the number of c-fos-positive cells. Our results indicate that extended positive fighting experience in a social conflict heightens aggression, increases proliferation of neuronal progenitors and production of young neurons in the hippocampus, and decreases neuronal activity in the amygdala; these changes can be modified by depriving the winners of the opportunity for further fights.

8.
Cell ; 158(3): 579-92, 2014 Jul 31.
Article in English | MEDLINE | ID: mdl-25083869

ABSTRACT

The p53 tumor suppressor coordinates a series of antiproliferative responses that restrict the expansion of malignant cells, and as a consequence, p53 is lost or mutated in the majority of human cancers. Here, we show that p53 restricts expression of the stem and progenitor-cell-associated protein nestin in an Sp1/3 transcription-factor-dependent manner and that Nestin is required for tumor initiation in vivo. Moreover, loss of p53 facilitates dedifferentiation of mature hepatocytes into nestin-positive progenitor-like cells, which are poised to differentiate into hepatocellular carcinomas (HCCs) or cholangiocarcinomas (CCs) in response to lineage-specific mutations that target Wnt and Notch signaling, respectively. Many human HCCs and CCs show elevated nestin expression, which correlates with p53 loss of function and is associated with decreased patient survival. Therefore, transcriptional repression of Nestin by p53 restricts cellular plasticity and tumorigenesis in liver cancer.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Nestin/metabolism , Animals , Carcinoma, Hepatocellular/pathology , Cell Transformation, Neoplastic , Hepatocytes/metabolism , Humans , Liver Neoplasms/pathology , Mice , Prognosis , Sp1 Transcription Factor/metabolism , Sp3 Transcription Factor/metabolism , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism
9.
PLoS One ; 9(3): e91762, 2014.
Article in English | MEDLINE | ID: mdl-24667609

ABSTRACT

Individuals exposed to social stress in childhood are more predisposed to developing psychoemotional disorders in adulthood. Here we use an animal model to determine the influence of hostile social environment in adolescence on behavior during adult life. One-month-old adolescent male mice were placed for 2 weeks in a common cage with an adult aggressive male. Animals were separated by a transparent perforated partition, but the adolescent male was exposed daily to short attacks from the adult male. After exposure to social stress, some of the adolescent mice were placed for 3 weeks in comfortable conditions. Following this rest period, stressed young males and adult males were studied in a range of behavioral tests to evaluate the levels of anxiety, depressiveness, and communicativeness with an unfamiliar partner. In addition, adult mice exposed to social stress in adolescence were engaged in agonistic interactions. We found that 2 weeks of social stress result in a decrease of communicativeness in the home cage and diminished social interactions on the novel territory. Stressed adolescents demonstrated a high level of anxiety in the elevated plus-maze test and helplessness in the Porsolt test. Furthermore, the number of dividing (BrdU-positive) cells in the subgranular zone of the dentate gyrus was significantly lower in stressed adolescents. After 3 weeks of rest, most behavioral characteristics in different tests, as well as the number of BrdU-positive cells in the hippocampus, did not differ from those of the respective control mice. However, the level of anxiety remained high in adult males exposed to chronic social stress in childhood. Furthermore, these males were more aggressive in the agonistic interactions. Thus, hostile social environment in adolescence disturbs psychoemotional state and social behaviors of animals in adult life.


Subject(s)
Aggression/psychology , Anxiety/psychology , Behavior, Animal , Social Behavior , Stress, Psychological/psychology , Animals , Chronic Disease , Hippocampus/cytology , Male , Maze Learning , Mice , Mice, Inbred C57BL , Social Environment , Stress, Psychological/physiopathology
10.
Proc Natl Acad Sci U S A ; 110(44): E4142-51, 2013 Oct 29.
Article in English | MEDLINE | ID: mdl-24127602

ABSTRACT

Learning related paradigms play an important role in shaping the development and specificity of synaptic networks, notably by regulating mechanisms of spine growth and pruning. The molecular events underlying these synaptic rearrangements remain poorly understood. Here we identify NO signaling as a key mediator of activity-dependent excitatory synapse development. We find that chronic blockade of NO production in vitro and in vivo interferes with the development of hippocampal and cortical excitatory spine synapses. The effect results from a selective loss of activity-mediated spine growth mechanisms and is associated with morphological and functional alterations of remaining synapses. These effects of NO are mediated by a cGMP cascade and can be reproduced or prevented by postsynaptic expression of vasodilator-stimulated phosphoprotein phospho-mimetic or phospho-resistant mutants. In vivo analyses show that absence of NO prevents the increase in excitatory synapse density induced by environmental enrichment and interferes with the formation of local clusters of excitatory synapses. We conclude that NO plays an important role in regulating the development of excitatory synapses by promoting local activity-dependent spine-growth mechanisms.


Subject(s)
Hippocampus/growth & development , Learning/physiology , Nerve Net/growth & development , Neuronal Plasticity/physiology , Nitric Oxide/metabolism , Signal Transduction/physiology , Synapses/physiology , Animals , Cell Adhesion Molecules/metabolism , Cyclic GMP/metabolism , DNA Primers/genetics , Evoked Potentials/physiology , Genotype , Mice , Microfilament Proteins/metabolism , Microscopy, Electron , Patch-Clamp Techniques , Phosphoproteins/metabolism , Phosphorylation , Polymerase Chain Reaction , Rats , Synapses/ultrastructure
11.
Eur J Neurosci ; 37(12): 1987-93, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23773068

ABSTRACT

Production of new neurons from stem cells is important for cognitive function, and the reduction of neurogenesis in the aging brain may contribute to the accumulation of age-related cognitive deficits. Restriction of calorie intake and prolonged treatment with rapamycin have been shown to extend the lifespan of animals and delay the onset of the age-related decline in tissue and organ function. Using a reporter line in which neural stem and progenitor cells are marked by the expression of green fluorescent protein (GFP), we examined the effect of prolonged exposure to calorie restriction (CR) or rapamycin on hippocampal neural stem and progenitor cell proliferation in aging mice. We showed that CR increased the number of dividing cells in the dentate gyrus of female mice. The majority of these cells corresponded to nestin-GFP-expressing neural stem or progenitor cells; however, this increased proliferative activity of stem and progenitor cells did not result in a significant increase in the number of doublecortin-positive newborn neurons. Our results suggest that restricted calorie intake may increase the number of divisions that neural stem and progenitor cells undergo in the aging brain of females.


Subject(s)
Aging/physiology , Caloric Restriction , Hippocampus/cytology , Neural Stem Cells/cytology , Neurogenesis/physiology , Aging/metabolism , Animals , Female , Male , Mice , Mice, Transgenic , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Sirolimus/pharmacology
12.
Nature ; 495(7440): 241-5, 2013 Mar 14.
Article in English | MEDLINE | ID: mdl-23467088

ABSTRACT

Epithelial ovarian cancer (EOC) is the fifth leading cause of cancer deaths among women in the United States, but its pathogenesis is poorly understood. Some epithelial cancers are known to occur in transitional zones between two types of epithelium, whereas others have been shown to originate in epithelial tissue stem cells. The stem cell niche of the ovarian surface epithelium (OSE), which is ruptured and regenerates during ovulation, has not yet been defined unequivocally. Here we identify the hilum region of the mouse ovary, the transitional (or junction) area between the OSE, mesothelium and tubal (oviductal) epithelium, as a previously unrecognized stem cell niche of the OSE. We find that cells of the hilum OSE are cycling slowly and express stem and/or progenitor cell markers ALDH1, LGR5, LEF1, CD133 and CK6B. These cells display long-term stem cell properties ex vivo and in vivo, as shown by our serial sphere generation and long-term lineage-tracing assays. Importantly, the hilum cells show increased transformation potential after inactivation of tumour suppressor genes Trp53 and Rb1, whose pathways are altered frequently in the most aggressive and common type of human EOC, high-grade serous adenocarcinoma. Our study supports experimentally the idea that susceptibility of transitional zones to malignant transformation may be explained by the presence of stem cell niches in those areas. Identification of a stem cell niche for the OSE may have important implications for understanding EOC pathogenesis.


Subject(s)
Epithelium/pathology , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/pathology , Ovary/pathology , Stem Cell Niche , Animals , Biomarkers/analysis , Biomarkers/metabolism , Carcinoma, Ovarian Epithelial , Cell Lineage , Cell Separation , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Epithelium/metabolism , Female , Male , Mice , Neoplasms, Glandular and Epithelial/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Ovarian Neoplasms/metabolism , Ovary/metabolism , Receptors, G-Protein-Coupled/metabolism , Retinoblastoma Protein/deficiency , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
13.
Cell Stem Cell ; 8(5): 566-79, 2011 May 06.
Article in English | MEDLINE | ID: mdl-21549330

ABSTRACT

Production of new neurons in the adult hippocampus decreases with age; this decline may underlie age-related cognitive impairment. Here we show that continuous depletion of the neural stem cell pool, as a consequence of their division, may contribute to the age-related decrease in hippocampal neurogenesis. Our results indicate that adult hippocampal stem cells, upon exiting their quiescent state, rapidly undergo a series of asymmetric divisions to produce dividing progeny destined to become neurons and subsequently convert into mature astrocytes. Thus, the decrease in the number of neural stem cells is a division-coupled process and is directly related to their production of new neurons. We present a scheme of the neurogenesis cascade in the adult hippocampus that includes a proposed "disposable stem cell" model and accounts for the disappearance of hippocampal neural stem cells, the appearance of new astrocytes, and the age-related decline in the production of new neurons.


Subject(s)
Aging/physiology , Astrocytes/metabolism , Cognition Disorders/pathology , Hippocampus/pathology , Neural Stem Cells/metabolism , Animals , Astrocytes/pathology , Cell Differentiation , Cell Division , Cell Line , Cell Survival , Cognition Disorders/physiopathology , Computational Biology , Green Fluorescent Proteins/genetics , Intermediate Filament Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nestin , Neural Stem Cells/pathology , Stem Cell Niche
14.
Cell Stem Cell ; 8(2): 164-76, 2011 Feb 04.
Article in English | MEDLINE | ID: mdl-21295273

ABSTRACT

The p53 homolog p63 is essential for development, yet its role in cancer is not clear. We discovered that p63 deficiency evokes the tumor-suppressive mechanism of cellular senescence, causing a striking absence of stratified epithelia such as the skin. Here we identify the predominant p63 isoform, ΔNp63α, as a protein that bypasses oncogene-induced senescence to drive tumorigenesis in vivo. Interestingly, bypass of senescence promotes stem-like proliferation and maintains survival of the keratin 15-positive stem cell population. Furthermore, we identify the chromatin-remodeling protein Lsh as a new target of ΔNp63α that is an essential mediator of senescence bypass. These findings indicate that ΔNp63α is an oncogene that cooperates with Ras to promote tumor-initiating stem-like proliferation and suggest that Lsh-mediated chromatin-remodeling events are critical to this process.


Subject(s)
DNA Helicases/metabolism , Phosphoproteins/metabolism , Skin/cytology , Stem Cells/cytology , Stem Cells/metabolism , Trans-Activators/metabolism , Animals , Cell Proliferation , Cells, Cultured , Chromatin Immunoprecipitation , Flow Cytometry , Humans , Keratinocytes/metabolism , Mice , Mice, Nude , Phosphoproteins/genetics , Polymerase Chain Reaction , Protein Binding , Trans-Activators/genetics
15.
Nature ; 466(7308): 829-34, 2010 Aug 12.
Article in English | MEDLINE | ID: mdl-20703299

ABSTRACT

The cellular constituents forming the haematopoietic stem cell (HSC) niche in the bone marrow are unclear, with studies implicating osteoblasts, endothelial and perivascular cells. Here we demonstrate that mesenchymal stem cells (MSCs), identified using nestin expression, constitute an essential HSC niche component. Nestin(+) MSCs contain all the bone-marrow colony-forming-unit fibroblastic activity and can be propagated as non-adherent 'mesenspheres' that can self-renew and expand in serial transplantations. Nestin(+) MSCs are spatially associated with HSCs and adrenergic nerve fibres, and highly express HSC maintenance genes. These genes, and others triggering osteoblastic differentiation, are selectively downregulated during enforced HSC mobilization or beta3 adrenoreceptor activation. Whereas parathormone administration doubles the number of bone marrow nestin(+) cells and favours their osteoblastic differentiation, in vivo nestin(+) cell depletion rapidly reduces HSC content in the bone marrow. Purified HSCs home near nestin(+) MSCs in the bone marrow of lethally irradiated mice, whereas in vivo nestin(+) cell depletion significantly reduces bone marrow homing of haematopoietic progenitors. These results uncover an unprecedented partnership between two distinct somatic stem-cell types and are indicative of a unique niche in the bone marrow made of heterotypic stem-cell pairs.


Subject(s)
Hematopoietic Stem Cells/cytology , Mesenchymal Stem Cells/cytology , Stem Cell Niche/cytology , Animals , Cell Differentiation/drug effects , Cell Division , Cell Lineage/drug effects , Cell Movement , Cells, Cultured , Chemokine CXCL12/metabolism , Chondrocytes/cytology , Chondrocytes/drug effects , Gene Expression Regulation/genetics , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Intermediate Filament Proteins/metabolism , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, Transgenic , Multipotent Stem Cells/cytology , Multipotent Stem Cells/drug effects , Multipotent Stem Cells/metabolism , Nerve Tissue Proteins/metabolism , Nestin , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Parathyroid Hormone/pharmacology , Stem Cell Niche/drug effects , Stem Cell Niche/metabolism , Stromal Cells/cytology , Stromal Cells/drug effects , Stromal Cells/metabolism , Sympathetic Nervous System/physiology
16.
Endocrinology ; 150(12): 5520-9, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19837875

ABSTRACT

Atrial (ANP) and C-type (CNP) natriuretic peptide generate physiological effects via selective activation of two closely related membrane receptors with guanylyl cyclase (GC) activity, known as GC-A and GC-B. As yet, however, the discrete roles for ANP/GC-A vs. CNP/GC-B signaling in many mammalian tissues are still poorly understood. We here used receptor affinity labeling and GC assays to characterize comparatively GC-A/GC-B expression and functional activity during rat brain development. The study revealed that GC-B predominates in the developing and GC-A in the adult brain, with regional differences each between cerebral cortex, cerebellum, and brain stem. Whereas GC-A levels nearly continuously increase between embryonal d 18 and adult, GC-B expression in brain is highest and widely distributed around postnatal d 1. The striking perinatal GC-B peak coincides with elevated expression of nestin, a marker protein for neural stem/progenitor cells. Immunohistochemical investigations revealed a cell body-restricted subcellular localization of GC-B and perinatal abundance of GC-B-expressing cells in regions high in nestin-expressing cells. However, and supported by examination of nestin-GFP transgenic mice, GC-B and nestin are not coexpressed in the same cells. Rather, GC-B(+) cells are distinguished by expression of NeuN, an early marker of differentiating neurons. These findings suggest that GC-B(+) cells represent neuronal fate-specific progeny of nestin(+) progenitors and raise the attention to specific and pronounced activities of CNP/GC-B signaling during perinatal brain maturation. The absence of this activity may cause the neurological disorders observed in GC-B-deficient mice.


Subject(s)
Brain/enzymology , Guanylate Cyclase/metabolism , Neurogenesis , Animals , Atrial Natriuretic Factor/metabolism , Brain/embryology , Brain/growth & development , Brain Stem/embryology , Brain Stem/growth & development , Brain Stem/metabolism , Cerebellum/embryology , Cerebellum/growth & development , Cerebellum/metabolism , Cerebral Cortex/embryology , Cerebral Cortex/growth & development , Cerebral Cortex/metabolism , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunoblotting , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/metabolism , Isoenzymes/metabolism , Male , Mice , Mice, Transgenic , Microscopy, Confocal , Microscopy, Fluorescence , Natriuretic Peptide, C-Type/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nestin , Photoaffinity Labels , Rats , Rats, Wistar , Time Factors
17.
Proc Natl Acad Sci U S A ; 105(17): 6332-7, 2008 Apr 29.
Article in English | MEDLINE | ID: mdl-18436641

ABSTRACT

Adult tissues undergo continuous cell turnover in response to stress, damage, or physiological demand. New differentiated cells are generated from dedicated or facultative stem cells or from self-renewing differentiated cells. Here we describe a different stem cell strategy for tissue maintenance, distinct from that observed for dedicated or facultative stem cells. We report the presence of nestin-expressing adult stem cells in the perilumenal region of the mature anterior pituitary and, using genetic inducible fate mapping, demonstrate that they serve to generate subsets of all six terminally differentiated endocrine cell types of the pituitary gland. These stem cells, while not playing a significant role in organogenesis, undergo postnatal expansion and start producing differentiated progeny, which colonize the organ that initially entirely consisted of differentiated cells derived from embryonic precursors. This generates a mosaic organ with two phenotypically similar subsets of endocrine cells that have different origins and different life histories. These parallel but distinct lineages of differentiated cells in the gland may help the maturing organism adapt to changes in the metabolic regulatory landscape.


Subject(s)
Adult Stem Cells/cytology , Pituitary Gland, Anterior/cytology , Adult Stem Cells/metabolism , Animals , Cell Differentiation , Cell Lineage , Cell Proliferation , Green Fluorescent Proteins/metabolism , Intermediate Filament Proteins/metabolism , Mice , Mice, Inbred C57BL , Models, Biological , Nerve Tissue Proteins/metabolism , Nestin , Pituitary Neoplasms/pathology
18.
Mol Med ; 14(3-4): 141-9, 2008.
Article in English | MEDLINE | ID: mdl-18091979

ABSTRACT

Nitric oxide (NO) signaling is important for the regulation of hematopoiesis. However, the role of individual NO synthase (NOS) isoforms is unclear. Our results indicate that the neuronal NOS isoform (nNOS) regulates hematopoiesis in vitro and in vivo. nNOS is expressed in adult bone marrow and fetal liver and is enriched in stromal cells. There is a strong correlation between expression of nNOS in a panel of stromal cell lines established from bone marrow and fetal liver and the ability of these cell lines to support hematopoietic stem cells; furthermore, NO donor can further increase this ability. The number of colonies generated in vitro from the bone marrow and spleen of nNOS-null mutants is increased relative to wild-type or inducible- or endothelial NOS knockout mice. These results describe a new role for nNOS beyond its action in the brain and muscle and suggest a model where nNOS, expressed in stromal cells, produces NO which acts as a paracrine regulator of hematopoietic stem cells.


Subject(s)
Hematopoiesis/physiology , Isoenzymes/metabolism , Nitric Oxide Synthase Type I/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/enzymology , Cells, Cultured , Female , Hepatocytes/cytology , Hepatocytes/enzymology , Isoenzymes/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type I/genetics , Paracrine Communication , Spleen/cytology , Spleen/enzymology , Stem Cells/cytology , Stem Cells/enzymology , Stromal Cells/cytology , Stromal Cells/enzymology
19.
Mol Cell Biol ; 27(15): 5430-44, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17515607

ABSTRACT

We describe a novel stress-induced gene, noxin, and a knockout mouse line with an inactivated noxin gene. The noxin gene does not have sequelogs in the genome and encodes a highly serine-rich protein with predicted phosphorylation sites for ATM, Akt, and DNA-dependent protein kinase kinases; nuclear localization signals; and a Zn finger domain. noxin mRNA and protein levels are under tight control by the cell cycle. noxin, identified as a nitric oxide-inducible gene, is strongly induced by a wide range of stress signals: gamma- and UV irradiation, hydrogen peroxide, adriamycin, and cytokines. This induction is dependent on p53. Noxin accumulates in the nucleus in response to stress and, when ectopically expressed, Noxin arrests the cell cycle at G1; although it also induces p53, the cell cycle arrest function of Noxin is independent of p53 activity. noxin knockout mice are viable and fertile; however, they have an enlarged heart, several altered hematopoietic parameters, and a decreased number of spermatids. Importantly, loss or downregulation of Noxin leads to increased cell death. Our results suggest that Noxin may be a component of the cell defense system: it is activated by various stress stimuli, helps cells to withdraw from cycling, and opposes apoptosis.


Subject(s)
Apoptosis , Carrier Proteins/genetics , Cell Cycle , Phosphoproteins/genetics , Amino Acid Sequence , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Cell Cycle/drug effects , Cell Cycle Proteins , Cloning, Molecular , Down-Regulation/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Profiling , Gene Targeting , Mice , Mice, Knockout , Molecular Sequence Data , NIH 3T3 Cells , Nitric Oxide/pharmacology , Organ Size/drug effects , Phenotype , Phosphoproteins/chemistry , Phosphoproteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , S-Nitroso-N-Acetylpenicillamine/pharmacology , Subcellular Fractions/metabolism , Tumor Suppressor Protein p53/genetics
20.
Dev Dyn ; 234(2): 413-21, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16127706

ABSTRACT

Oval cells, which become apparent in the liver after chronic injury, serve as bipotent progenitors for differentiated hepatocytes and cholangiocytes. We found that, in the liver of adult transgenic mice in which expression of green fluorescent protein (GFP) is driven by regulatory elements of the nestin gene, the GFP signal marks a subpopulation of small epithelial cells that meet the criteria for oval cells, including morphology, localization, antigenic profile, and reactivity in response to injury. In the regenerating and developing liver, we also found nestin-GFP-positive cells that express hepatocyte markers; such cells may correspond to transiently appearing differentiating progeny of oval cells. During development, GFP-expressing cells in the liver emerge relatively late, after the appearance of differentiated hepatocytes and cholangiocytes. Our results suggest that nestin-GFP cells in the liver correspond to a specialized cell type whose primary function may be to serve as a reserve for adult liver epithelial cell types.


Subject(s)
Biomarkers , Green Fluorescent Proteins/metabolism , Intermediate Filament Proteins/biosynthesis , Intermediate Filament Proteins/genetics , Liver/metabolism , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Animals , Cell Adhesion Molecules/metabolism , Cell Lineage , Enhancer Elements, Genetic , Epithelial Cells/metabolism , Epithelium/metabolism , Hepatocytes/metabolism , Immunohistochemistry , Intermediate Filament Proteins/metabolism , Liver/embryology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Nerve Tissue Proteins/metabolism , Nestin , Stem Cells/metabolism , alpha-Fetoproteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...