Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Chem Rev ; 123(3): 1166-1205, 2023 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-36696538

RESUMO

Mass cytometry (cytometry by time-of-flight detection [CyTOF]) is a bioanalytical technique that enables the identification and quantification of diverse features of cellular systems with single-cell resolution. In suspension mass cytometry, cells are stained with stable heavy-atom isotope-tagged reagents, and then the cells are nebulized into an inductively coupled plasma time-of-flight mass spectrometry (ICP-TOF-MS) instrument. In imaging mass cytometry, a pulsed laser is used to ablate ca. 1 µm2 spots of a tissue section. The plume is then transferred to the CyTOF, generating an image of biomarker expression. Similar measurements are possible with multiplexed ion bean imaging (MIBI). The unit mass resolution of the ICP-TOF-MS detector allows for multiparametric analysis of (in principle) up to 130 different parameters. Currently available reagents, however, allow simultaneous measurement of up to 50 biomarkers. As new reagents are developed, the scope of information that can be obtained by mass cytometry continues to increase, particularly due to the development of new small molecule reagents which enable monitoring of active biochemistry at the cellular level. This review summarizes the history and current state of mass cytometry reagent development and elaborates on areas where there is a need for new reagents. Additionally, this review provides guidelines on how new reagents should be tested and how the data should be presented to make them most meaningful to the mass cytometry user community.


Assuntos
Indicadores e Reagentes , Biomarcadores/análise
2.
Can Vet J ; 59(9): 993-996, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30197443

RESUMO

The echocardiographic findings of a young Pomeranian-cross dog with tetralogy of Fallot, patent foramen ovale, and tricuspid valve dysplasia are described. Ongoing medical management of hypoxemia and erythrocytosis was carried out and the dog survived to 2 years of age. Treatment options for tetralogy of Fallot are discussed.


Tétralogie de Fallot avec persistance du foramen ovale et dysplasie de la valve tricuspide concomitante chez un chien. Les constatations écho-cardiographiques chez un chien Poméranien de race croisée atteint de la tétralogie de Fallot, de la persistance du foramen ovale et d'une dysplasie de la valve tricuspide sont décrites. Une gestion médicale constante de l'hypoxémie et de l'érythrocytose a été réalisée et le chien a survécu jusqu'à l'âge de 2 ans. Les options de traitement pour la tétralogie de Fallot sont discutées.(Traduit par Isabelle Vallières).


Assuntos
Doenças do Cão/congênito , Forame Oval Patente/veterinária , Tetralogia de Fallot/veterinária , Animais , Cães , Ecocardiografia/métodos , Ecocardiografia/veterinária , Forame Oval Patente/diagnóstico por imagem , Hipóxia/terapia , Hipóxia/veterinária , Masculino , Policitemia/terapia , Policitemia/veterinária , Tetralogia de Fallot/diagnóstico por imagem , Tetralogia de Fallot/terapia , Valva Tricúspide/anormalidades
3.
Am J Hum Genet ; 92(5): 725-43, 2013 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-23643382

RESUMO

Congenital hypogonadotropic hypogonadism (CHH) and its anosmia-associated form (Kallmann syndrome [KS]) are genetically heterogeneous. Among the >15 genes implicated in these conditions, mutations in FGF8 and FGFR1 account for ~12% of cases; notably, KAL1 and HS6ST1 are also involved in FGFR1 signaling and can be mutated in CHH. We therefore hypothesized that mutations in genes encoding a broader range of modulators of the FGFR1 pathway might contribute to the genetics of CHH as causal or modifier mutations. Thus, we aimed to (1) investigate whether CHH individuals harbor mutations in members of the so-called "FGF8 synexpression" group and (2) validate the ability of a bioinformatics algorithm on the basis of protein-protein interactome data (interactome-based affiliation scoring [IBAS]) to identify high-quality candidate genes. On the basis of sequence homology, expression, and structural and functional data, seven genes were selected and sequenced in 386 unrelated CHH individuals and 155 controls. Except for FGF18 and SPRY2, all other genes were found to be mutated in CHH individuals: FGF17 (n = 3 individuals), IL17RD (n = 8), DUSP6 (n = 5), SPRY4 (n = 14), and FLRT3 (n = 3). Independently, IBAS predicted FGF17 and IL17RD as the two top candidates in the entire proteome on the basis of a statistical test of their protein-protein interaction patterns to proteins known to be altered in CHH. Most of the FGF17 and IL17RD mutations altered protein function in vitro. IL17RD mutations were found only in KS individuals and were strongly linked to hearing loss (6/8 individuals). Mutations in genes encoding components of the FGF pathway are associated with complex modes of CHH inheritance and act primarily as contributors to an oligogenic genetic architecture underlying CHH.


Assuntos
Fosfatase 6 de Especificidade Dupla/genética , Fatores de Crescimento de Fibroblastos/genética , Predisposição Genética para Doença/genética , Hipogonadismo/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana/genética , Proteínas do Tecido Nervoso/genética , Receptores de Interleucina/genética , Algoritmos , Animais , Sequência de Bases , Biologia Computacional , Feminino , Estudos de Associação Genética , Humanos , Imuno-Histoquímica , Padrões de Herança/genética , Masculino , Glicoproteínas de Membrana , Camundongos , Dados de Sequência Molecular , Mutação/genética , Análise de Sequência de DNA , Homologia de Sequência , Ressonância de Plasmônio de Superfície
4.
Behav Brain Funct ; 11(1): 34, 2015 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-26537115

RESUMO

BACKGROUND: Fibroblast growth factors (FGFs) are crucial signaling molecules that direct the development of the vertebrate brain. FGF8 gene signaling in particular, may be important for the development of the hypothalamus-pituitary-adrenal (HPA)-axis. Indeed, newborn Fgf8 hypomorphic mice harbor a major reduction in the number of vasopressin (VP) neurons in the paraventricular nucleus (PVN), the central output component of the HPA-axis. Additionally, recent studies indicated that adult heterozygous ((+/neo)) Fgf8 hypomorphic mice exhibit more anxiety-like behaviors than wildtype (WT) mice. These studies led us to investigate whether Fgf8 hypomorphy abrogated VP and/or corticotropin-releasing hormone (CRH) neuronal development in the postnatal day (PN) 21 and adult mouse PVN. Furthermore, we studied whether Fgf8 hypomorphy disrupted HPA responsiveness in these mice. METHODS: Using immunohistochemistry, we examined the development of VP and CRH neurons located in the PVN of PN 21 and adult Fgf8 (+/neo) mice. Moreover, we used a restraint stress (RS) paradigm and measured corticosterone levels with enzyme immunoassays in order to assess HPA axis activation. RESULTS: The number of VP neurons in the PVN did not differ between WT and Fgf8 (+/neo) mice on PN 21 and in adulthood. In contrast, CRH immunoreactivity was much higher in Fgf8 (+/neo) mice than in WT mice on PN 21, this difference was no longer shown in adult mice. RS caused a higher increase in corticosterone levels in adult Fgf8 (+/neo) mice than in WT mice after 15 min, but no difference was seen after 45 min. CONCLUSIONS: First, Fgf8 hypomorphy did not eliminate VP and CRH neurons in the mouse PVN, but rather disrupted the postnatal timing of neuropeptide expression onset in PVN neurons. Second, Fgf8 hypomorphy may, in part, be an explanation for affective disorders involving hyperactivity of the HPA axis, such as anxiety.


Assuntos
Fator 8 de Crescimento de Fibroblasto/fisiologia , Células Neuroendócrinas/fisiologia , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/crescimento & desenvolvimento , Animais , Contagem de Células , Corticosterona/sangue , Hormônio Liberador da Corticotropina/metabolismo , Fator 8 de Crescimento de Fibroblasto/genética , Sistema Hipotálamo-Hipofisário/fisiologia , Masculino , Camundongos , Camundongos Transgênicos , Células Neuroendócrinas/citologia , Sistema Hipófise-Suprarrenal/fisiologia , Restrição Física , Vasopressinas/metabolismo
5.
Pflugers Arch ; 465(5): 573-84, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23503727

RESUMO

The concept that the brain differs in make-up between males and females is not new. For example, it is well established that anatomists in the nineteenth century found sex differences in human brain weight. The importance of sex differences in the organization of the brain cannot be overstated as they may directly affect cognitive functions, such as verbal skills and visuospatial tasks in a sex-dependent fashion. Moreover, the incidence of neurological and psychiatric diseases is also highly dependent on sex. These clinical observations reiterate the importance that gender must be taken into account as a relevant possible contributing factor in order to understand the pathogenesis of neurological and psychiatric disorders. Gender-dependent differentiation of the brain has been detected at every level of organization--morphological, neurochemical, and functional--and has been shown to be primarily controlled by sex differences in gonadal steroid hormone levels during perinatal development. In this review, we discuss howthe gonadal steroid hormone testosterone and its metabolites affect downstream signaling cascades, including gonadal steroid receptor activation, and epigenetic events in order to differentiate the brain in a gender-dependent fashion.


Assuntos
Encéfalo/crescimento & desenvolvimento , Epigênese Genética , Caracteres Sexuais , Animais , Encéfalo/metabolismo , Encéfalo/fisiologia , Feminino , Hormônios Esteroides Gonadais/metabolismo , Humanos , Masculino
6.
Front Endocrinol (Lausanne) ; 14: 1166132, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37181038

RESUMO

Neurons that secrete gonadotropin-releasing hormone (GnRH) drive vertebrate reproduction. Genetic lesions that disrupt these neurons in humans lead to congenital hypogonadotropic hypogonadism (CHH) and reproductive failure. Studies on CHH have largely focused on the disruption of prenatal GnRH neuronal migration and postnatal GnRH secretory activity. However, recent evidence suggests a need to also focus on how GnRH neurons initiate and maintain their identity during prenatal and postnatal periods. This review will provide a brief overview of what is known about these processes and several gaps in our knowledge, with an emphasis on how disruption of GnRH neuronal identity can lead to CHH phenotypes.


Assuntos
Hormônio Liberador de Gonadotropina , Hipogonadismo , Humanos , Hormônio Liberador de Gonadotropina/genética , Hipogonadismo/genética , Hipogonadismo/congênito , Neurônios , Movimento Celular , Fenótipo
7.
Am J Physiol Endocrinol Metab ; 303(12): E1428-39, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23047985

RESUMO

The continued presence of gonadotropin-releasing hormone (GnRH) neurons is required for a healthy reproductive lifespan, but factors that maintain postnatal GnRH neurons have not been identified. To begin to understand these factors, we investigated whether 1) fibroblast growth factor (FGF) signaling and 2) interactions with the opposite sex are involved in the maintenance of the postnatal GnRH system. A transgenic mouse model (dnFGFR mouse) with the targeted expression of a dominant-negative FGF receptor (dnFGFR) in GnRH neurons was used to examine the consequence of FGF signaling deficiency on postnatal GnRH neurons. Male dnFGFR mice suffered a significant loss of postnatal GnRH neurons within the first 100 days of life. Interestingly, this loss was reversed after cohabitation with female, but not male, mice for 300-550 days. Along with a rescue in GnRH neuron numbers, opposite-sex housing in dnFGFR males also increased hypothalamic GnRH peptide levels, promoted a more mature GnRH neuronal morphology, facilitated litter production, and enhanced testicular morphology. Last, mice hypomorphic for FGFR3 exhibited a similar pattern of postnatal GnRH neuronal loss as dnFGFR males, suggesting FGF signaling acts, in part, through FGFR3 to enhance the maintenance of the postnatal GnRH system. In summary, we have shown that FGF signaling is required for the continued presence of postnatal GnRH neurons. However, this requirement is not absolute, since sexual interactions can compensate for defects in FGFR signaling, thereby rescuing the declining GnRH system. This suggests the postnatal GnRH system is highly plastic and capable of responding to environmental stimuli throughout adult life.


Assuntos
Envelhecimento , Fator 3 de Crescimento de Fibroblastos/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Neurônios/metabolismo , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais , Animais , Contagem de Células , Heterozigoto , Hipotálamo/citologia , Hipotálamo/crescimento & desenvolvimento , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Degeneração Neural/etiologia , Degeneração Neural/prevenção & controle , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética , Receptores LHRH/metabolismo , Comportamento Sexual Animal , Transmissão Sináptica , Testículo/citologia , Testículo/crescimento & desenvolvimento , Testículo/metabolismo
8.
Front Neuroendocrinol ; 32(1): 95-107, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21129392

RESUMO

Fibroblast growth factor (FGF) signaling is pivotal to the formation of numerous central regions. Increasing evidence suggests FGF signaling also directs the development of the neuroendocrine hypothalamus, a collection of neuroendocrine neurons originating primarily within the nose and the ventricular zone of the diencephalon. This review outlines evidence for a role of FGF signaling in the prenatal and postnatal development of several hypothalamic neuroendocrine systems. The emphasis is placed on the nasally derived gonadotropin-releasing hormone neurons, which depend on neurotrophic cues from FGF signaling throughout the neurons' lifetime. Although less is known about neuroendocrine neurons derived from the diencephalon, recent studies suggest they also exhibit variable levels of dependence on FGF signaling. Overall, FGF signaling provides a broad spectrum of cues that ranges from genesis, cell survival/death, migration, morphological changes, to hormone synthesis in the neuroendocrine hypothalamus. Abnormal FGF signaling will deleteriously impact multiple hypothalamic neuroendocrine systems, resulting in the disruption of diverse physiological functions.


Assuntos
Fatores de Crescimento de Fibroblastos/fisiologia , Hipotálamo/embriologia , Sistemas Neurossecretores/embriologia , Animais , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Hipotálamo/metabolismo , Modelos Biológicos , Sistemas Neurossecretores/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/genética , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
9.
Biol Reprod ; 86(4): 119, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22278983

RESUMO

Fibroblast growth factor (FGF) signaling is essential for the development of the gonadotropin-releasing hormone (GnRH) system. Mice harboring deficiencies in Fgf8 or Fgf receptor 1 (Fgfr1) suffer a significant loss of GnRH neurons, but their reproductive phenotypes have not been examined. This study examined if female mice hypomorphic for Fgf8, Fgfr1, or both (compound hypomorphs) exhibited altered parameters of pubertal onset, estrous cyclicity, and fertility. Further, we examined the number of kisspeptin (KP)-immunoreactive (ir) neurons in the anteroventral periventricular/periventricular nuclei (AVPV/PeV) of these mice to assess if changes in the KP system, which stimulates the GnRH system, could contribute to the reproductive phenotypes. Single hypomorphs (Fgfr1(+/-) or Fgf8(+/-)) had normal timing for vaginal opening (VO) but delayed first estrus. However, after achieving the first estrus, they underwent normal expression of estrous cycles. In contrast, the compound hypomorphs underwent early VO and normal first estrus, but had disorganized estrous cycles that subsequently reduced their fertility. KP immunohistochemistry on Postnatal Day 15, 30, and 60 transgenic female mice revealed that female compound hypomorphs had significantly more KP-ir neurons in the AVPV/PeV compared to their wild-type littermates, suggesting increased KP-ir neurons may drive early VO but could not maintain the cyclic changes in GnRH neuronal activity required for female fertility. Overall, these data suggest that Fgf signaling deficiencies differentially alter the parameters of female pubertal onset and cyclicity. Further, these deficiencies led to changes in the AVPV/PeV KP-ir neurons that may have contributed to the accelerated VO in the compound hypomorphs.


Assuntos
Fator 8 de Crescimento de Fibroblasto/metabolismo , Infertilidade Feminina/metabolismo , Kisspeptinas/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Reprodução/fisiologia , Animais , Núcleos Anteriores do Tálamo/metabolismo , Comunicação Celular/fisiologia , Ciclo Estral/metabolismo , Feminino , Fator 8 de Crescimento de Fibroblasto/genética , Hormônio Liberador de Gonadotropina/metabolismo , Camundongos , Camundongos Transgênicos , Núcleos da Linha Média do Tálamo/metabolismo , Neurônios/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Maturidade Sexual/fisiologia , Transdução de Sinais/fisiologia
10.
Behav Brain Funct ; 8: 47, 2012 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-22950531

RESUMO

BACKGROUND: Fibroblast growth factors (FGFs) and their receptors (FGFRs) are necessary for the proper development of gonadotropin-releasing hormone (GnRH) neurons, which are key activators of the hypothalamo-pituitary-gonadal axis. Transgenic mice that have the targeted expression of a dominant negative FGFR (dnFGFR) in GnRH neurons (dnFGFR mice) have a 30% decrease of GnRH neurons. Additionally, only 30-40% of the pups born to the transgenic dams survive to weaning age. These data raised the possibility that FGFR defects in GnRH neurons could adversely affect maternal behavior via novel mechanisms. METHODS: We first determined if defective maternal behavior in dnFGFR mothers may contribute to poor pup survival by measuring pup retrieval and a battery of maternal behaviors in primiparous control (n=10-12) and dnFGFR (n=13-14) mothers. Other endocrine correlates of maternal behaviors, including plasma estradiol levels and hypothalamic pro-oxyphysin and GnRH transcript levels were also determined using enzyme-linked immunoassay and quantitative reverse transcription polymerase chain reaction, respectively. RESULTS: Maternal behaviors (% time crouching with pups, time off pups but not feeding, time feeding, and total number of nesting bouts) were not significantly different in dnFGFR mice. However, dnFGFR dams were more likely to leave their pups scattered and took significantly longer to retrieve each pup compared to control dams. Further, dnFGFR mothers had significantly lower GnRH transcripts and circulating E2, but normal pro-oxyphysin transcript levels. CONCLUSIONS: Overall, this study suggests a complex scenario in which a GnRH system compromised by reduced FGF signaling leads to not only suboptimal reproductive physiology, but also suboptimal maternal behavior.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Comportamento Materno/fisiologia , Neurônios/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Animais , Comportamento Animal/fisiologia , Estradiol/sangue , Hormônio Liberador de Gonadotropina/genética , Camundongos , Camundongos Transgênicos , Receptores de Fatores de Crescimento de Fibroblastos/genética
11.
J Clin Invest ; 118(8): 2822-31, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18596921

RESUMO

Idiopathic hypogonadotropic hypogonadism (IHH) with anosmia (Kallmann syndrome; KS) or with a normal sense of smell (normosmic IHH; nIHH) are heterogeneous genetic disorders associated with deficiency of gonadotropin-releasing hormone (GnRH). While loss-of-function mutations in FGF receptor 1 (FGFR1) cause human GnRH deficiency, to date no specific ligand for FGFR1 has been identified in GnRH neuron ontogeny. Using a candidate gene approach, we identified 6 missense mutations in FGF8 in IHH probands with variable olfactory phenotypes. These patients exhibited varied degrees of GnRH deficiency, including the rare adult-onset form of hypogonadotropic hypogonadism. Four mutations affected all 4 FGF8 splice isoforms (FGF8a, FGF8b, FGF8e, and FGF8f), while 2 mutations affected FGF8e and FGF8f isoforms only. The mutant FGF8b and FGF8f ligands exhibited decreased biological activity in vitro. Furthermore, mice homozygous for a hypomorphic Fgf8 allele lacked GnRH neurons in the hypothalamus, while heterozygous mice showed substantial decreases in the number of GnRH neurons and hypothalamic GnRH peptide concentration. In conclusion, we identified FGF8 as a gene implicated in GnRH deficiency in both humans and mice and demonstrated an exquisite sensitivity of GnRH neuron development to reductions in FGF8 signaling.


Assuntos
Fator 8 de Crescimento de Fibroblasto/metabolismo , Hormônio Liberador de Gonadotropina/deficiência , Transdução de Sinais , Adulto , Animais , Estudos de Casos e Controles , Estudos de Coortes , Feminino , Fator 8 de Crescimento de Fibroblasto/química , Fator 8 de Crescimento de Fibroblasto/genética , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Heterozigoto , Humanos , Hipogonadismo/genética , Hipogonadismo/fisiopatologia , Síndrome de Kallmann/genética , Síndrome de Kallmann/fisiopatologia , Masculino , Camundongos , Camundongos Transgênicos , Modelos Moleculares , Mutação , Neurônios/citologia , Neurônios/metabolismo , Transtornos do Olfato/genética , Linhagem
12.
Endocrinology ; 162(9)2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34147032

RESUMO

The rapid decline of circulating 17ß-estradiol (E2) at menopause leads to negative neurological consequences, although hormone therapy paradoxically has both harmful and positive effects depending on the age at which it is delivered. The inconsistent response to E2 suggests unappreciated regulatory mechanisms for estrogen receptors (ERs), and we predicted it could be due to age-related differences in ERß phosphorylation. We assessed ERß phosphorylation using a sensitive mass spectrometry approach that provides absolute quantification (AQUA-MS) of individually phosphorylated residues. Specifically, we quantified phosphorylated ERß in the hippocampus of women (aged 21-83 years) and in a rat model of menopause at 4 residues with conserved sequence homology between the 2 species: S105, S176, S200, and Y488. Phosphorylation at these sites, which spanned all domains of ERß, were remarkably consistent between the 2 species, showing high levels of S105 phosphorylation (80%-100%) and low levels of S200 (20%-40%). Further, S200 phosphorylation decreased with aging in humans and loss of E2 in rats. Surprisingly, Y488 phosphorylation, which has been linked to ERß ligand-independent actions, exhibited approximately 70% phosphorylation, unaltered by species, age, or E2, suggesting ERß's primary mode of action may not require E2 binding. We further show phosphorylation at 2 sites directly altered ERß DNA-binding efficiency, and thus could affect its transcription factor activity. These findings provide the first absolute quantification of ERß phosphorylation in the human and rat brain, novel insights into ERß regulation, and a critical foundation for providing more targeted therapeutic options for menopause in the future.


Assuntos
Receptor beta de Estrogênio/análise , Hipocampo/química , Menopausa/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Envelhecimento/metabolismo , Envelhecimento/patologia , Aminoácidos/análise , Aminoácidos/metabolismo , Animais , Estradiol/análise , Estradiol/metabolismo , Receptor beta de Estrogênio/metabolismo , Feminino , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Pessoa de Meia-Idade , Modelos Animais , Fragmentos de Peptídeos/análise , Fragmentos de Peptídeos/metabolismo , Fosforilação , Ratos , Ratos Endogâmicos F344 , Adulto Jovem
13.
Artigo em Inglês | MEDLINE | ID: mdl-33800199

RESUMO

Understanding the sedentary patterns can guide the design of strategies to engage older adults in physical activity. This scoping review aimed to synthesize available evidence on sedentary behaviors in care facilities. We searched PubMed/MEDLINE and Web of Science for studies published from inception through October 2020. Eighteen studies were included and reviewed according to the Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) guidelines. Data obtained were analyzed based on levels of care provided. Overall, daily sedentary time was higher among residents in high level care facilities (e.g., nursing homes) (11.6 h/day) than intermediate/mixed level care facilities (e.g., assisted living) (9.5 h/day). In intermediate/mixed level care facilities, television (TV) viewing was the most common sedentary activity (2.5-2.9 h/day; 26% of daily sedentary time), while napping was the most favorite sedentary activity (4.7 h/day; 36% of waking hours) in high level care facilities. Sex differences in daily patterns of sedentary behavior (sedentary time, uninterrupted bouts, and bout durations) were commonly observed in intermediate/mixed level care facilities, as exemplified by men being more sedentary by 0.7-1.1 h/day. In summary, this study highlights distinctive sedentary patterns among older adults residing in different levels of care facilities, addressing a pressing need for customized interventions to engage care facility residents in physical activity.


Assuntos
Exercício Físico , Comportamento Sedentário , Idoso , Atenção à Saúde , Feminino , Humanos , Masculino , Casas de Saúde
14.
Artigo em Inglês | MEDLINE | ID: mdl-34567220

RESUMO

Most women live with an inactive lifestyle, which suggests a need for preference-based choices to promote their participation in physical activity. This systematic review synthesized key findings on the health benefits of Qigong among women. We conducted a systematic search of randomized controlled trials (RCTs) of Qigong among women according to the PRISMA guidelines using the following databases from their inception through March 2021: PubMed/MEDLINE, Web of Science, Cochrane Library, and US National Library of Medicine. The risk of bias was examined using the Cochrane Collaboration's tool for assessing the risk of bias in randomized trials. Altogether, 18 RCTs were included for final review. Results showed that Qigong was a feasible exercise in improving health outcomes, particularly depressive symptoms (63% of trials), quality of life (43%), and fatigue (29%), among general women, intimate partner violence survivors, and women with chronic conditions (e.g., breast cancer patients or survivors). Almost 90% (7/8) of trials reported high adherence rates ranging from 73 to 95% for supervised group training and 63 to 80% for home self-practice. Thus far, there was no evidence of serious adverse effects from performing Qigong. For the risk of bias across trials, a lack of allocation concealment (72% of trials), no blinding of participants and personnel (67%), and incomplete outcome data (67%) were the major sources. In summary, Qigong is a safe, feasible, and beneficial exercise for general women, abused sufferers, and health-compromised women. However, given the potential risk of bias found in many studies, improved rigor of study design in future trials will be imperatively required.

15.
J Neuroendocrinol ; 32(6): e12860, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32452569

RESUMO

Mammalian reproductive success depends on gonadotrophin-releasing hormone (GnRH) neurones to stimulate gonadotrophin secretion from the anterior pituitary and activate gonadal steroidogenesis and gametogenesis. Genetic screening studies in patients diagnosed with Kallmann syndrome (KS), a congenital form of hypogonadotrophic hypogonadism (CHH), identified several causal mutations, including those in the fibroblast growth factor (FGF) system. This signalling pathway regulates neuroendocrine progenitor cell proliferation, fate specification and cell survival. Indeed, the GnRH neurone system was absent or abrogated in transgenic mice with reduced (ie, hypomorphic) Fgf8 and/or Fgf receptor (Fgfr) 1 expression, respectively. Moreover, we found that GnRH neurones were absent in the embryonic olfactory placode of Fgf8 hypomorphic mice, the putative birthplace of GnRH neurones. These observations, together with those made in human KS/CHH patients, indicate that the FGF8/FGFR1 signalling system is a requirement for the ontogenesis of the GnRH neuronal system and function. In this review, we discuss how epigenetic factors control the expression of genes such as Fgf8 that are known to be critical for GnRH neurone ontogenesis, fate specification, and the pathogenesis of KS/CHH.


Assuntos
Epigênese Genética/fisiologia , Hipogonadismo/genética , Neurogênese/genética , Neurônios/fisiologia , Animais , Epigenômica , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Humanos , Hipogonadismo/patologia , Hipogonadismo/psicologia , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Neurônios/patologia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais/fisiologia
16.
Neurosci Lett ; 714: 134569, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31644920

RESUMO

Down syndrome is the most common genetic cause of intellectual disability and occurs due to the trisomy of human chromosome 21. Adolescent and adult brains from humans with Down syndrome exhibit various neurological phenotypes including a reduction in the size of the corpus callosum, hippocampal commissure and anterior commissure. However, it is unclear when and how these interhemispheric connectivity defects arise. Using the Ts65Dn mouse model of Down syndrome, we examined interhemispheric connectivity in postnatal day 0 (P0) Ts65Dn mouse brains. We find that there is no change in the volume of the corpus callosum or anterior commissure in P0 Ts65Dn mice. However, the volume of the hippocampal commissure is significantly reduced in P0 Ts65Dn mice, and this may contribute to the impaired learning and memory phenotype of this disorder. Interhemispheric connectivity defects that arise during development may be due to disrupted axon growth. In line with this, we find that developing hippocampal neurons display reduced axon length in vitro, as compared to neurons from their euploid littermates. This study is the first to report the presence of defective interhemispheric connectivity at the time of birth in Ts65Dn mice, providing evidence that early therapeutic intervention may be an effective time window for the treatment of Down syndrome.


Assuntos
Comissura Anterior/patologia , Axônios/patologia , Corpo Caloso/patologia , Síndrome de Down/patologia , Fórnice/patologia , Animais , Animais Recém-Nascidos , Comissura Anterior/fisiopatologia , Orientação de Axônios/fisiologia , Tamanho Celular , Corpo Caloso/fisiopatologia , Modelos Animais de Doenças , Síndrome de Down/fisiopatologia , Fórnice/fisiopatologia , Hipocampo/patologia , Hipocampo/fisiopatologia , Técnicas In Vitro , Camundongos , Camundongos Transgênicos , Vias Neurais , Neurogênese/fisiologia , Crescimento Neuronal , Neurônios/patologia , Tamanho do Órgão
17.
PLoS One ; 14(7): e0220530, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31361780

RESUMO

Fibroblast growth factor 8 (FGF8) is a potent morphogen that regulates the ontogenesis of gonadotropin-releasing hormone (GnRH) neurons, which control the hypothalamus-pituitary-gonadal (HPG) axis, and therefore reproductive success. Indeed, FGF8 and FGFR1 deficiency severely compromises vertebrate reproduction in mice and humans and is associated with Kallmann Syndrome (KS), a congenital disease characterized by hypogonadotropic hypogonadism associated with anosmia. Our laboratory demonstrated that FGF8 signaling through FGFR1, both of which are KS-related genes, is necessary for proper GnRH neuron development in mice and humans. Here, we investigated the possibility that non-genetic factors, such as the epigenome, may contribute to KS onset. For this purpose, we developed an embryonic explant model, utilizing the mouse olfactory placode (OP), the birthplace of GnRH neurons. We show that TET1, which converts 5-methylcytosine residues (5mC) to 5-hydroxymethylated cytosines (5hmC), controls transcription of Fgf8 during GnRH neuron ontogenesis. Through MeDIP and ChIP RT-qPCR we found that TET1 bound to specific CpG islands on the Fgf8 promoter. We found that the temporal expression of Fgf8 correlates with not only TET1 binding, but also with 5hmC enrichment. siRNA knockdown of Tet1 reduced Fgf8 and Fgfr1 mRNA expression. During this time period, Fgf8 also switched histone status, most likely via recruitment of EZH2, a major component of the polycomb repressor complex-2 (PRC2) at E13.5. Together, these studies underscore the significance of epigenetics and chromatin modifications to temporally regulated genes involved in KS.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Fator 8 de Crescimento de Fibroblasto/genética , Regulação da Expressão Gênica , Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transcrição Gênica , Animais , Células Cultivadas , Proteínas de Ligação a DNA/genética , Feminino , Histonas/genética , Histonas/metabolismo , Masculino , Camundongos , Neurônios/citologia , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas/genética , Transdução de Sinais
18.
Endocrinology ; 149(10): 4997-5003, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18566132

RESUMO

GnRH neurons are essential for the onset and maintenance of reproduction. Mutations in both fibroblast growth factor receptor (Fgfr1) and Fgf8 have been shown to cause Kallmann syndrome, a disease characterized by hypogonadotropic hypogonadism and anosmia, indicating that FGF signaling is indispensable for the formation of a functional GnRH system. Presently it is unclear which stage of GnRH neuronal development is most impacted by FGF signaling deficiency. GnRH neurons express both FGFR1 and -3; thus, it is also unclear whether FGFR1 or FGFR3 contributes directly to GnRH system development. In this study, we examined the developing GnRH system in mice deficient in FGF8, FGFR1, or FGFR3 to elucidate the individual contribution of these FGF signaling components. Our results show that the early emergence of GnRH neurons from the embryonic olfactory placode requires FGF8 signaling, which is mediated through FGFR1, not FGFR3. These data provide compelling evidence that the developing GnRH system is exquisitely sensitive to reduced levels of FGF signaling. Furthermore, Kallmann syndrome stemming from FGF signaling deficiency may be due primarily to defects in early GnRH neuronal development prior to their migration into the forebrain.


Assuntos
Fator 8 de Crescimento de Fibroblasto/metabolismo , Hormônio Liberador de Gonadotropina/fisiologia , Condutos Olfatórios/embriologia , Condutos Olfatórios/fisiologia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais/fisiologia , Animais , Apoptose/fisiologia , Movimento Celular/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Hipotálamo/citologia , Hipotálamo/embriologia , Hipotálamo/fisiologia , Proteínas de Filamentos Intermediários/metabolismo , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/metabolismo , Neurônios/fisiologia , Condutos Olfatórios/citologia , Periferinas , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo , Transativadores/metabolismo , Órgão Vomeronasal/citologia , Órgão Vomeronasal/embriologia , Órgão Vomeronasal/fisiologia
19.
Endocrinology ; 148(7): 3371-82, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17412808

RESUMO

Arginine vasopressin (AVP) is a neuropeptide involved in the regulation of fluid balance, stress, circadian rhythms, and social behaviors. In the brain, AVP is tightly regulated by gonadal steroid hormones in discrete regions with gonadectomy abolishing and testosterone replacement restoring normal AVP expression in adult males. Previous studies demonstrated that 17beta-estradiol, a primary metabolite of testosterone, is responsible for restoring most of the AVP expression in the brain after castration. However, 5alpha-dihydrotestosterone (DHT) has also been shown to play a role in the regulation of AVP expression, thus implicating the involvement of both androgen and estrogen receptors (ER). Furthermore, DHT, through its conversion to 5alpha-androstane-3beta,17beta-diol, has been shown to modulate estrogen response element-mediated promoter activity through an ER pathway. The present study addressed two central hypotheses: 1) that androgens directly modulate AVP promoter activity and 2) the effect is mediated by an estrogen or androgen receptor pathway. To that end, we overexpressed androgen receptor, ERbeta, and ERbeta splice variants in a neuronal cell line and measured AVP promoter activity using a firefly luciferase reporter assay. Our results demonstrate that DHT and its metabolite 5alpha-androstane-3beta,17beta-diol stimulate AVP promoter activity through ERbeta in a neuronal cell line.


Assuntos
Arginina Vasopressina/genética , Di-Hidrotestosterona/farmacologia , Receptor beta de Estrogênio/genética , Neurônios/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Androgênios/química , Androgênios/metabolismo , Androstano-3,17-diol/farmacologia , Arginina Vasopressina/metabolismo , Sequência de Bases , Linhagem Celular Tumoral , Receptor beta de Estrogênio/metabolismo , Expressão Gênica/efeitos dos fármacos , Vetores Genéticos/genética , Humanos , Imuno-Histoquímica , Luciferases/genética , Luciferases/metabolismo , Dados de Sequência Molecular , Estrutura Molecular , Mutagênese Sítio-Dirigida , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
J Comp Neurol ; 505(3): 249-67, 2007 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-17879269

RESUMO

Estrogens regulate neural processes such as neuronal development, reproductive behavior, and hormone secretion, and signal through estrogen receptor (ER) alpha and ERbeta (here called ERbeta1). Recent studies have found variations in ERalpha and ERbeta1 mRNA splicing in rodents and humans. Functional reporter gene assays suggest that these splicing variations alter ER-mediated transcriptional regulation. Estrogen receptor beta 2 (ERbeta2), an ERbeta1 splice variant containing an 18 amino acid (AA) insert in the ligand binding domain, binds estradiol with approximately 10-fold lower affinity than ERbeta1, suggesting that it may serve as a low-affinity ER. Moreover, ERbeta2 reportedly acts in a dominant-negative fashion when heterodimerized with ERbeta1 or ERalpha. To explore the function of ERbeta2 in brain, an antiserum (TwobetaER.1) targeting the 18 AA insert was developed and characterized. Western blot analysis and transient expression of ERbeta2 in cell lines demonstrated that TwobetaER.1 recognizes ERbeta2. In the adult female rat brain, ERbeta2 immunoreactivity is localized in the cell nucleus and is expressed with a distribution similar to that of ERbeta1 mRNA. ERbeta2 immunoreactive cell numbers were high in, for example, piriform cortex, paraventricular nucleus, supraoptic nucleus, arcuate nucleus, and hippocampal CA regions, whereas it was low in the dentate gyrus. Moreover, ERbeta2 is coexpressed in gonadotropin-releasing hormone and oxytocin neurons. These studies demonstrate ERbeta splice variant proteins in brain and support the hypothesis that ER signaling diversity depends not only on ligand or coregulatory proteins, but also on regional and phenotypic selectivity of ER splice variant proteins.


Assuntos
Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Mesencéfalo/metabolismo , Prosencéfalo/metabolismo , Processamento Alternativo , Animais , Western Blotting , Feminino , Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/metabolismo , Ovariectomia , Ocitocina/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA