Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Physiol Genomics ; 56(3): 265-275, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38145289

ABSTRACT

Agouti-related peptide (AgRP/Agrp) within the hypothalamic arcuate nucleus (ARC) contributes to the control of energy balance, and dysregulated Agrp may contribute to metabolic adaptation during prolonged obesity. In mice, three isoforms of Agrp are encoded via distinct first exons. Agrp-A (ENSMUST00000005849.11) contributed 95% of total Agrp in mouse ARC, whereas Agrp-B (ENSMUST00000194654.2) dominated in placenta (73%). Conditional deletion of Klf4 from Agrp-expressing cells (Klf4Agrp-KO mice) reduced Agrp mRNA and increased energy expenditure but had no effects on food intake or the relative abundance of Agrp isoforms in the ARC. Chronic high-fat diet feeding masked these effects of Klf4 deletion, highlighting the context-dependent contribution of KLF4 to Agrp control. In the GT1-7 mouse hypothalamic cell culture model, which expresses all three isoforms of Agrp (including Agrp-C, ENSMUST00000194091.6), inhibition of extracellular signal-regulated kinase (ERK) simultaneously increased KLF4 binding to the Agrp promoter and stimulated Agrp expression. In addition, siRNA-mediated knockdown of Klf4 reduced expression of Agrp. We conclude that the expression of individual isoforms of Agrp in the mouse is dependent upon cell type and that KLF4 directly promotes the transcription of Agrp via a mechanism that is superseded during obesity.NEW & NOTEWORTHY In mice, three distinct isoforms of Agouti-related peptide are encoded via distinct first exons. In the arcuate nucleus of the hypothalamus, Krüppel-like factor 4 stimulates transcription of the dominant isoform in lean mice, but this mechanism is altered during diet-induced obesity.


Subject(s)
Agouti-Related Protein , Kruppel-Like Factor 4 , Neurons , Animals , Mice , Agouti-Related Protein/genetics , Agouti-Related Protein/metabolism , Hypothalamus/metabolism , Neurons/metabolism , Obesity/genetics , Obesity/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism
2.
Physiol Genomics ; 54(6): 196-205, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35476598

ABSTRACT

The brain renin-angiotensin system (RAS) is implicated in control of blood pressure (BP), fluid intake, and energy expenditure (EE). Angiotensin II (ANG II) within the arcuate nucleus of the hypothalamus contributes to control of resting metabolic rate (RMR) and thereby EE through its actions on Agouti-related peptide (AgRP) neurons, which also contribute to EE control by leptin. First, we determined that although leptin stimulates EE in control littermates, mice with transgenic activation of the brain RAS (sRA) exhibit increased EE and leptin has no additive effect to exaggerate EE in these mice. These findings led us to hypothesize that leptin and ANG II in the brain stimulate EE through a shared mechanism. Because AgRP signaling to the melanocortin MC4R receptor contributes to the metabolic effects of leptin, we performed a series of studies examining RMR, fluid intake, and BP responses to ANG II in mice rendered deficient for expression of MC4R via a transcriptional block (Mc4r-TB). These mice were resistant to stimulation of RMR in response to activation of the endogenous brain RAS via chronic deoxycorticosterone acetate (DOCA)-salt treatment, whereas fluid and electrolyte effects remained intact. These mice were also resistant to stimulation of RMR via acute intracerebroventricular (ICV) injection of ANG II, whereas BP responses to ICV ANG II remained intact. Collectively, these data demonstrate that the effects of ANG II within the brain to control RMR and EE are dependent on MC4R signaling, whereas fluid homeostasis and BP responses are independent of MC4R signaling.


Subject(s)
Angiotensin II , Energy Metabolism , Leptin , Receptor, Melanocortin, Type 4 , Agouti-Related Protein/metabolism , Angiotensin II/pharmacology , Animals , Blood Pressure/physiology , Brain/metabolism , Energy Metabolism/physiology , Leptin/metabolism , Leptin/pharmacology , Melanocortins/metabolism , Melanocortins/pharmacology , Mice , Receptor, Melanocortin, Type 4/metabolism
3.
Am J Physiol Regul Integr Comp Physiol ; 322(6): R467-R485, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35348007

ABSTRACT

Hypertension characterized by low circulating renin activity accounts for roughly 25%-30% of primary hypertension in humans and can be modeled experimentally via deoxycorticosterone acetate (DOCA)-salt treatment. In this model, phenotypes develop in progressive phases, although the timelines and relative contributions of various mechanisms to phenotype development can be distinct between laboratories. To explore interactions among environmental influences such as diet formulation and dietary sodium (Na) content on phenotype development in the DOCA-salt paradigm, we examined an array of cardiometabolic endpoints in young adult male C57BL/6J mice during sham or DOCA-salt treatments when mice were maintained on several common, commercially available laboratory rodent "chow" diets including PicoLab 5L0D (0.39% Na), Envigo 7913 (0.31% Na), Envigo 2920x (0.15% Na), or a customized version of Envigo 2920x (0.4% Na). Energy balance (weight gain, food intake, digestive efficiency, and energy efficiency), fluid and electrolyte homeostasis (fluid intake, Na intake, fecal Na content, hydration, and fluid compartmentalization), renal functions (urine production rate, glomerular filtration rate, urine Na excretion, renal expression of renin, vasopressin receptors, aquaporin-2 and relationships among markers of vasopressin release, aquaporin-2 shedding, and urine osmolality), and blood pressure, all exhibited changes that were subject to interactions between diet and DOCA-salt. Interestingly, some of these phenotypes, including blood pressure and hydration, were dependent on nonsodium dietary components, as Na-matched diets resulted in distinct phenotype development. These findings provide a broad and robust illustration of an environment × treatment interaction that impacts the use and interpretation of a common rodent model of low-renin hypertension.


Subject(s)
Desoxycorticosterone Acetate , Hypertension , Animals , Aquaporin 2 , Blood Pressure/physiology , Desoxycorticosterone/pharmacology , Desoxycorticosterone Acetate/pharmacology , Diet , Hypertension/metabolism , Male , Mice , Mice, Inbred C57BL , Renin/metabolism , Sodium/metabolism
4.
Am J Physiol Regul Integr Comp Physiol ; 323(4): R410-R421, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35816717

ABSTRACT

The renin-angiotensin system (RAS) within the brain is implicated in the control of fluid and electrolyte balance, autonomic functions, blood pressure, and energy expenditure. Mouse models are increasingly used to explore these mechanisms; however, sex and dose dependencies of effects elicited by chronic intracerebroventricular (ICV) angiotensin II (ANG II) infusion have not been carefully established in this species. To examine the interactions among sex, body mass, and ICV ANG II on ingestive behaviors and energy balance, young adult C57BL/6J mice of both sexes were studied in a multiplexed metabolic phenotyping system (Promethion) during chronic infusion of ANG II (0, 5, 20, or 50 ng/h). At these infusion rates, ANG II caused accelerating dose-dependent increases in drinking and total energy expenditure in male mice, but female mice exhibited a complex biphasic response with maximum responses at 5 ng/h. Body mass differences did not account for sex-dependent differences in drinking behavior or total energy expenditure. In contrast, resting metabolic rate was similarly increased by ICV ANG II in a dose-dependent manner in both sexes after correction for body mass. We conclude that chronic ICV ANG II stimulates water intake, resting, and total energy expenditure in male C57BL/6J mice following straightforward accelerating dose-dependent kinetics, but female C57BL/6J mice exhibit complex biphasic responses to ICV ANG II. Furthermore, control of resting metabolic rate by ANG II is dissociable from mechanisms controlling fluid intake and total energy expenditure. Future studies of the sex dependency of ANG II within the brain of mice must be designed to carefully consider the biphasic responses that occur in females.


Subject(s)
Angiotensin II , Angiotensin II/pharmacology , Animals , Blood Pressure/physiology , Female , Homeostasis , Infusions, Intraventricular , Injections, Intraventricular , Male , Mice , Mice, Inbred C57BL
5.
Am J Physiol Regul Integr Comp Physiol ; 320(4): R438-R451, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33439766

ABSTRACT

Postnatal growth failure is a common morbidity for preterm infants and is associated with adverse neurodevelopmental outcomes. Although sodium (Na) deficiency early in life impairs somatic growth, its impact on neurocognitive functions has not been extensively studied. We hypothesized that Na deficiency during early life is sufficient to cause growth failure and program neurobehavioral impairments in later life. C57BL/6J mice were placed on low- (0.4), normal- (1.5), or high- (3 g/kg) Na chow at weaning (PD22) and continued on the diet for 3 wk (to PD40). Body composition and fluid distribution were determined serially by time-domain NMR and bioimpedance spectroscopy, and anxiety, learning, and memory were assessed using the elevated plus maze and Morris water maze paradigms in later adulthood (PD63-PD69). During the diet intervention, body mass gains were suppressed in the low- compared with normal- and high-Na groups despite similar caloric uptake rates across groups. Fat mass was reduced in males but not in females fed low-Na diet. Fat-free mass and hydration were significantly reduced in both males and females fed the low-Na diet, although rapidly corrected after return to normal diet. Measures of anxiety-like behavior and learning in adulthood were not affected by diet in either sex, yet memory performance was modified by a complex interaction between sex and early life Na intake. These data support the concepts that Na deficiency impairs growth and that the amount of Na intake which supports optimal somatic growth during early life may be insufficient to fully support neurocognitive development.


Subject(s)
Behavior, Animal , Diet, Sodium-Restricted/adverse effects , Nervous System/growth & development , Nutritional Status , Sodium, Dietary/administration & dosage , Spatial Memory , Water-Electrolyte Balance , Age Factors , Animal Feed , Animal Nutritional Physiological Phenomena , Animals , Body Composition , Elevated Plus Maze Test , Female , Male , Mice, Inbred C57BL , Morris Water Maze Test , Weight Gain
6.
Am J Physiol Regul Integr Comp Physiol ; 320(1): R44-R54, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33085913

ABSTRACT

The measurement of fluid compartmentalization, or the distribution of fluid volume between extracellular (ECF) and intracellular (ICF) spaces, historically requires complicated, burdensome, and often terminal methodologies that do not permit repeated or longitudinal experiments. New technologies including time-domain nuclear magnetic resonance (TD-NMR)-based methods allow for highly accurate measurements of total body water (TBW) within minutes in a noninvasive manner, but do not permit dissection of ECF versus ICF reservoirs. In contrast, methods such as bioimpedance spectroscopy (BIS) allow dissection of ECF versus ICF reservoirs but are hampered by dependence on many nuanced details in data collection that undermine confidence in experimental results. Here, we present a novel combinatorial use of these two technologies (NMR/BIS) to improve the accuracy of BIS-based assessments of ECF and ICF, while maintaining the advantages of these minimally invasive methods. Briefly, mice undergo TD-NMR and BIS-based measures, and then fat masses as derived by TD-NMR are used to correct BIS outputs. Mice of the C57BL/6J background were studied using NMR/BIS methods to assess the effects of acute furosemide injection and diet-induced obesity on fluid compartmentalization, and to examine the influence of sex, body mass and composition, and diet on TBW, ECF, and ICF. We discovered that in mice, sex and body size/composition have substantial and interactive effects on fluid compartmentalization. We propose that the combinatorial use of NMR/BIS methods will enable a revisioning of the types of longitudinal, kinetic studies that can be performed to understand the impact of various interventions on body fluid homeostasis.


Subject(s)
Body Composition , Body Fluid Compartments/metabolism , Fluid Shifts , Magnetic Resonance Spectroscopy , Adiposity , Animals , Body Size , Electric Impedance , Female , Male , Mice, Inbred C57BL , Reproducibility of Results , Sex Factors
7.
Neuroendocrinology ; 111(3): 237-248, 2021.
Article in English | MEDLINE | ID: mdl-32335554

ABSTRACT

INTRODUCTION: Hyponatremia due to elevated arginine vasopressin (AVP) secretion increases mortality in liver failure patients. No previous studies have addressed sex differences in hyponatremia in liver failure animal models. OBJECTIVE: This study addressed this gap in our understanding of the potential sex differences in hyponatremia associated with increased AVP secretion. METHODS: This study tested the role of sex in the development of hyponatremia using adult male, female, and ovariectomized (OVX) female bile duct-ligated (BDL) rats. RESULTS: All BDL rats had significantly increased liver to body weight ratios compared to sham controls. Male BDL rats had hyponatremia with significant increases in plasma copeptin and FosB expression in supraoptic AVP neurons compared to male shams (all p < 0.05; 5-7). Female BDL rats did not become hyponatremic or demonstrate increased supraoptic AVP neuron activation and copeptin secretion compared to female shams. Plasma oxytocin was significantly higher in female BDL rats compared to female sham (p < 0.05; 6-10). This increase was not observed in male BDL rats. Ovariectomy significantly decreased plasma estradiol in sham rats compared to intact female sham (p < 0.05; 6-10). However, circulating estradiol was significantly elevated in OVX BDL rats compared to the OVX and female shams (p < 0.05; 6-10). Adrenal estradiol, testosterone, and dehydroepiandrosterone (DHEA) were measured to identify a possible source of circulating estradiol in OVX BDL rats. The OVX BDL rats had significantly increased adrenal estradiol along with significantly decreased adrenal testosterone and DHEA compared to OVX shams (all p < 0.05; 6-7). Plasma osmolality, hematocrit, copeptin, and AVP neuron activation were not significantly different between OVX BDL and OVX shams. Plasma oxytocin was significantly higher in OVX BDL rats compared to OVX sham. CONCLUSIONS: Our results show that unlike male BDL rats, female and OVX BDL rats did not develop hyponatremia, supraoptic AVP neuron activation, or increased copeptin secretion compared to female shams. Adrenal estradiol might have compensated for the lack of ovarian estrogens in OVX BDL rats.


Subject(s)
Arginine Vasopressin/metabolism , Bile Ducts , Estradiol/metabolism , Glycopeptides/metabolism , Hyponatremia/metabolism , Oxytocin/metabolism , Sex Characteristics , Supraoptic Nucleus/metabolism , Animals , Bile Ducts/surgery , Dehydroepiandrosterone/metabolism , Disease Models, Animal , Estradiol/blood , Female , Ligation , Male , Ovariectomy , Oxytocin/blood , Rats , Rats, Sprague-Dawley , Sex Factors , Testosterone/metabolism
8.
Am J Physiol Regul Integr Comp Physiol ; 318(5): R855-R869, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32186897

ABSTRACT

Angiotensin II (ANG II) Agtr1a receptor (AT1A) is expressed in cells of the arcuate nucleus of the hypothalamus that express the leptin receptor (Lepr) and agouti-related peptide (Agrp). Agtr1a expression in these cells is required to stimulate resting energy expenditure in response to leptin and high-fat diets (HFDs), but the mechanism activating AT1A signaling by leptin remains unclear. To probe the role of local paracrine/autocrine ANG II generation and signaling in this mechanism, we bred mice harboring a conditional allele for angiotensinogen (Agt, encoding AGT) with mice expressing Cre-recombinase via the Lepr or Agrp promoters to cause cell-specific deletions of Agt (AgtLepr-KO and AgtAgrp-KO mice, respectively). AgtLepr-KO mice were phenotypically normal, arguing against a paracrine/autocrine AGT signaling mechanism for metabolic control. In contrast, AgtAgrp-KO mice exhibited reduced preweaning survival, and surviving adults exhibited altered renal structure and steroid flux, paralleling previous reports of animals with whole body Agt deficiency or Agt disruption in albumin (Alb)-expressing cells (thought to cause liver-specific disruption). Surprisingly, adult AgtAgrp-KO mice exhibited normal circulating AGT protein and hepatic Agt mRNA expression but reduced Agt mRNA expression in adrenal glands. Reanalysis of RNA-sequencing data sets describing transcriptomes of normal adrenal glands suggests that Agrp and Alb are both expressed in this tissue, and fluorescent reporter gene expression confirms Cre activity in adrenal gland of both Agrp-Cre and Alb-Cre mice. These findings lead to the iconoclastic conclusion that extrahepatic (i.e., adrenal) expression of Agt is critically required for normal renal development and survival.


Subject(s)
Adrenal Glands/metabolism , Agouti-Related Protein/metabolism , Angiotensinogen/metabolism , Arcuate Nucleus of Hypothalamus/metabolism , Energy Metabolism , Kidney/metabolism , Receptors, Leptin/metabolism , Adrenal Glands/growth & development , Agouti-Related Protein/deficiency , Agouti-Related Protein/genetics , Angiotensinogen/deficiency , Angiotensinogen/genetics , Animals , Arcuate Nucleus of Hypothalamus/growth & development , Autocrine Communication , Female , Gene Expression Regulation, Developmental , Kidney/growth & development , Male , Mice, Knockout , Myocardium/metabolism , Paracrine Communication , Receptors, Leptin/deficiency , Receptors, Leptin/genetics , Serum Albumin/genetics , Serum Albumin/metabolism , Signal Transduction
9.
Neuroendocrinology ; 110(7-8): 630-641, 2020.
Article in English | MEDLINE | ID: mdl-31557760

ABSTRACT

Hyponatremia due to elevated arginine vasopressin (AVP) secretion increases mortality in liver failure patients. The mechanisms causing dysregulation of AVP secretion are unknown. Our hypothesis is that inappropriate AVP release associated with liver failure is due to increased brain-derived neurotrophic factor (BDNF) in the supraoptic nucleus (SON). BDNF diminishes GABAA inhibition in SON AVP neurons by increasing intracellular chloride through tyrosine receptor kinase B (TrkB) activation and downregulation of K+/Cl- cotransporter 2 (KCC2). This loss of inhibition could increase AVP secretion. This hypothesis was tested using shRNA against BDNF (shBDNF) in the SON in bile duct ligated (BDL) male rats. All BDL rats had significantly increased liver weight (p < 0.05; 6-9) compared to shams. BDL rats with control -shRNA injections (BDL scrambled [SCR]) developed hyponatremia with increased plasma AVP and copeptin (CPP; all p < 0.05; 6-9) compared to sham groups. This is the first study to show that phosphorylation of TrkB is significantly increased along with significant decrease in phosphorylation of KCC2 in BDL SCR rats compared to the sham rats (p < 0.05;6-8). Knockdown of BDNF in the SON of BDL rats (BDL shBDNF) significantly increased plasma osmolality and hematocrit compared to BDL SCR rats (p < 0.05; 6-9). The BDL shBDNF rats had significant (p < 0.05; 6-9) decreases in plasma AVP and CPP concentration compared to BDL SCR rats. The BDNF knockdown also significantly blocked the increase in TrkB phosphorylation and decrease in KCC2 phosphorylation (p < 0.05; 6-8). The results indicate that BDNF produced in the SON contributes to increased AVP secretion and hyponatremia during liver failure.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Hyponatremia/metabolism , Neurons/metabolism , Supraoptic Nucleus/metabolism , Vasopressins/metabolism , Animals , Disease Models, Animal , Hyponatremia/pathology , Liver Failure/metabolism , Liver Failure/pathology , Male , Neurons/pathology , Rats , Supraoptic Nucleus/pathology
10.
Am J Physiol Regul Integr Comp Physiol ; 315(5): R972-R982, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30156863

ABSTRACT

The median preoptic nucleus (MnPO) is an integrative site involved in body fluid homeostasis, cardiovascular control, thermoregulation, and sleep homeostasis. Angiotensin II (ANG II), a neuropeptide shown to have excitatory effects on MnPO neurons, is of particular interest with regard to its role in body fluid homeostasis and cardiovascular control. The present study investigated the role of angiotensin type 1a (AT1a) receptor activation on neuronal excitability in the MnPO. Male Sprague-Dawley rats were infused with an adeno-associated virus with an shRNA against the AT1a receptor or a scrambled control. In vitro loose-patch voltage-clamp recordings of spontaneous action potential activity were made from labeled MnPO neurons in response to brief focal application of ANG II or the GABAA receptor agonist muscimol. Additionally, tissue punches from MnPO were taken to asses mRNA and protein expression. AT1a receptor knockdown neurons were insensitive to ANG II and showed a marked reduction in GABAA-mediated inhibition. The reduction in GABAA-mediated inhibition was not associated with reductions in mRNA or protein expression of GABAA ß-subunits. Knockdown of the AT1a receptor was associated with a reduction in the potassium-chloride cotransporter KCC2 mRNA as well as a reduction in pS940 KCC2 protein. The impaired GABAA-mediated inhibition in AT1a knockdown neurons was recovered by bath application of phospholipase C and protein kinase C activators. The following study indicates that AT1a receptor activation mediates the excitability of MnPO neurons, in part, through the regulation of KCC2. The regulation of KCC2 influences the intracellular [Cl-] and the subsequent efficacy of GABAA-mediated currents.


Subject(s)
GABA-A Receptor Agonists/pharmacology , Receptor, Angiotensin, Type 1/drug effects , Receptors, GABA-A/drug effects , Symporters/metabolism , Action Potentials/physiology , Animals , Homeostasis/drug effects , Homeostasis/physiology , Male , Neurons/drug effects , Neurons/metabolism , Preoptic Area/drug effects , Rats, Sprague-Dawley , Receptors, GABA-A/metabolism , K Cl- Cotransporters
11.
Cell Rep ; 42(8): 112935, 2023 08 29.
Article in English | MEDLINE | ID: mdl-37540598

ABSTRACT

Resting metabolic rate (RMR) adaptation occurs during obesity and is hypothesized to contribute to failed weight management. Angiotensin II (Ang-II) type 1 (AT1A) receptors in Agouti-related peptide (AgRP) neurons contribute to the integrative control of RMR, and deletion of AT1A from AgRP neurons causes RMR adaptation. Extracellular patch-clamp recordings identify distinct cellular responses of individual AgRP neurons from lean mice to Ang-II: no response, inhibition via AT1A and Gαi, or stimulation via Ang-II type 2 (AT2) receptors and Gαq. Following diet-induced obesity, a subset of Ang-II/AT1A-inhibited AgRP neurons undergo a spontaneous G-protein "signal switch," whereby AT1A stop inhibiting the cell via Gαi and instead begin stimulating the cell via Gαq. DREADD-mediated activation of Gαi, but not Gαq, in AT1A-expressing AgRP cells stimulates RMR in lean and obese mice. Thus, loss of AT1A-Gαi coupling within the AT1A-expressing AgRP neuron subtype represents a molecular mechanism contributing to RMR adaptation.


Subject(s)
Neurons , Obesity , Receptor, Angiotensin, Type 1 , Animals , Mice , Agouti-Related Protein/metabolism , Angiotensin II/metabolism , Neurons/metabolism , Obesity/metabolism , Receptor, Angiotensin, Type 1/metabolism
12.
Sci Adv ; 9(48): eadg8118, 2023 12.
Article in English | MEDLINE | ID: mdl-38039359

ABSTRACT

Syncytiotrophoblast stress is theorized to drive development of preeclampsia, but its molecular causes and consequences remain largely undefined. Multiple hormones implicated in preeclampsia signal via the Gαq cascade, leading to the hypothesis that excess Gαq signaling within the syncytiotrophoblast may contribute. First, we present data supporting increased Gαq signaling and antioxidant responses within villous and syncytiotrophoblast samples of human preeclamptic placenta. Second, Gαq was activated in mouse placenta using Cre-lox and DREADD methodologies. Syncytiotrophoblast-restricted Gαq activation caused hypertension, kidney damage, proteinuria, elevated circulating proinflammatory factors, decreased placental vascularization, diminished spiral artery diameter, and augmented responses to mitochondrial-derived superoxide. Administration of the mitochondrial-targeted antioxidant Mitoquinone attenuated maternal proteinuria, lowered circulating inflammatory and anti-angiogenic mediators, and maintained placental vascularization. These data demonstrate a causal relationship between syncytiotrophoblast stress and the development of preeclampsia and identify elevated Gαq signaling and mitochondrial reactive oxygen species as a cause of this stress.


Subject(s)
Pre-Eclampsia , Animals , Mice , Pregnancy , Female , Humans , Trophoblasts , Placenta , Antioxidants/pharmacology , GTP-Binding Proteins , Proteinuria
13.
Front Oncol ; 12: 945689, 2022.
Article in English | MEDLINE | ID: mdl-36203435

ABSTRACT

Malignant tumors of the digestive system are common worldwide; however, it is extremely rare for more than two malignancies to occur simultaneously. Here, we report a case with a triple malignancy of the digestive system, including gastric, rectal, and hepatic tumors. The patient underwent surgical resection of three tumors followed by chemotherapy. Negative image-based screenings and the absence of serum tumor biomarkers elevation were found at 2.5 years after the surgery, indicating the absence of recurrence and metastasis of cancers.

14.
Front Synaptic Neurosci ; 14: 910820, 2022.
Article in English | MEDLINE | ID: mdl-35844900

ABSTRACT

Acute regulation of CO2 and pH homeostasis requires sensory feedback from peripheral (carotid body) and central (central) CO2/pH sensitive cells - so called respiratory chemoreceptors. Subsets of brainstem serotonin (5-HT) neurons in the medullary raphe are CO2 sensitive or insensitive based on differences in embryonic origin, suggesting these functionally distinct subpopulations may have unique transcriptional profiles. Here, we used Patch-to-Seq to determine if the CO2 responses in brainstem 5-HT neurons could be correlated to unique transcriptional profiles and/or unique molecular markers and pathways. First, firing rate changes with hypercapnic acidosis were measured in fluorescently labeled 5-HT neurons in acute brainstem slices from transgenic, Dahl SS (SSMcwi) rats expressing T2/ePet-eGFP transgene in Pet-1 expressing (serotonin) neurons (SS ePet1-eGFP rats). Subsequently, the transcriptomic and pathway profiles of CO2 sensitive and insensitive 5-HT neurons were determined and compared by single cell RNA (scRNAseq) and bioinformatic analyses. Low baseline firing rates were a distinguishing feature of CO2 sensitive 5-HT neurons. scRNAseq of these recorded neurons revealed 166 differentially expressed genes among CO2 sensitive and insensitive 5-HT neurons. Pathway analyses yielded novel predicted upstream regulators, including the transcription factor Egr2 and Leptin. Additional bioinformatic analyses identified 6 candidate gene markers of CO2 sensitive 5-HT neurons, and 2 selected candidate genes (CD46 and Iba57) were both expressed in 5-HT neurons determined via in situ mRNA hybridization. Together, these data provide novel insights into the transcriptional control of cellular chemoreception and provide unbiased candidate gene markers of CO2 sensitive 5-HT neurons. Methodologically, these data highlight the utility of the patch-to-seq technique in enabling the linkage of gene expression to specific functions, like CO2 chemoreception, in a single cell to identify potential mechanisms underlying functional differences in otherwise similar cell types.

15.
Hypertension ; 79(12): 2843-2853, 2022 12.
Article in English | MEDLINE | ID: mdl-36259376

ABSTRACT

BACKGROUND: RGS (regulator of G protein signaling) family members catalyze the termination of G protein signaling cascades. Single nucleotide polymorphisms in the RGS2 gene in humans have been linked to hypertension, preeclampsia, and anxiety disorders. Mice deficient for Rgs2 (Rgs2Null) exhibit hypertension, anxiety, and altered adipose development and function. METHODS: To study cell-specific functions of RGS2, a novel gene-targeted mouse harboring a conditional allele for the Rgs2 gene (Rgs2Flox) was developed. These mice were bred with mice expressing Cre-recombinase via the Agouti-related peptide locus (Agrp-Cre) to cause deletion of Rgs2 from all cells expressing Agrp (Rgs2Agrp-KO), or a novel transgenic mouse expressing Cre-recombinase via the ANG (angiotensin) type 1A receptor (Agtr1a/ AT1A) promoter encoded in a bacterial artificial chromosome (BAC-AT1A-Cre) to delete Rgs2 in all Agtr1a-expressing cells (Rgs2AT1A-KO). RESULTS: Whereas Rgs2Flox, Rgs2Agrp-KO, and BAC-AT1A-Cre mice exhibited normal growth and survival, Rgs2AT1A-KO exhibited pre-weaning lethality. Relative to littermates, Rgs2Agrp-KO exhibited reduced fat gains when maintained on a high fat diet, associated with increased energy expenditure. Similarly, surviving adult Rgs2AT1A-KO mice also exhibited increased energy expenditure. Surprisingly, given the hypertensive phenotype previously reported for Rgs2Null mice and evidence supporting a role for RGS2 in terminating AT1A signaling in various cell types, Rgs2AT1A-KO mice exhibited normal blood pressure, ingestive behaviors, and renal functions, both before and after chronic infusion of ANG (490 ng/kg/min, sc). CONCLUSIONS: These results demonstrate the development of a novel mouse with conditional expression of Rgs2 and illustrate the role of Rgs2 within selected cell types for cardiometabolic control.


Subject(s)
Hypertension , RGS Proteins , Animals , Mice , Agouti-Related Protein , Hypertension/genetics , Mice, Knockout , Mice, Transgenic , Receptor, Angiotensin, Type 1/genetics , Recombinases , RGS Proteins/genetics
16.
Hypertension ; 77(2): 420-431, 2021 02.
Article in English | MEDLINE | ID: mdl-33249862

ABSTRACT

Activation of central AT1Rs (angiotensin type 1 receptors) is required for the increased blood pressure, polydipsia, and salt intake in deoxycorticosterone acetate (DOCA)-salt hypertension. TRV120027 (TRV027) is an AT1R-biased agonist that selectively acts through ß-arrestin. We hypothesized that intracerebroventricular administration of TRV027 would ameliorate the effects of DOCA-salt. In a neuronal cell line, TRV027 induced AT1aR internalization through dynamin and clathrin-mediated endocytosis. We next evaluated the effect of chronic intracerebroventricular infusion of TRV027 on fluid intake. We measured the relative intake of water versus various saline solutions using a 2-bottle choice paradigm in mice subjected to DOCA with a concomitant intracerebroventricular infusion of either vehicle, TRV027, or losartan. Sham mice received intracerebroventricular vehicle without DOCA. TRV027 potentiated DOCA-induced water intake in the presence or absence of saline. TRV027 and losartan both increased the aversion for saline-an effect particularly pronounced for highly aversive saline solutions. Intracerebroventricular Ang (angiotensin) II, but not TRV027, increased water and saline intake in the absence of DOCA. In a separate cohort, blood pressure responses to acute intracerebroventricular injection of vehicle, TRV, or losartan were measured by radiotelemetry in mice with established DOCA-salt hypertension. Central administration of intracerebroventricular TRV027 or losartan each caused a significant and similar reduction of blood pressure and heart rate. We conclude that administration of TRV027 a selective ß-arrestin biased agonist directly into the brain increases aversion to saline and lowers blood pressure in a model of salt-sensitive hypertension. These data suggest that selective activation of AT1R ß-arrestin pathways may be exploitable therapeutically.


Subject(s)
Blood Pressure/drug effects , Choice Behavior/drug effects , Desoxycorticosterone/pharmacology , Hypertension/chemically induced , Neurons/drug effects , Receptor, Angiotensin, Type 1/agonists , beta-Arrestins/agonists , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Cell Line , Hypertension/metabolism , Losartan/pharmacology , Mice , Neurons/metabolism , Oligopeptides/pharmacology , Renin-Angiotensin System/drug effects
17.
Hypertension ; 76(2): 589-597, 2020 08.
Article in English | MEDLINE | ID: mdl-32507042

ABSTRACT

Prolonged obesity is associated with blunted feeding and thermogenic autonomic responses to leptin, but cardiovascular responses to leptin are maintained. This state of selective leptin resistance is, therefore, proposed to contribute to the pathogenesis and maintenance of obesity-associated hypertension. Cells of the arcuate nucleus of the hypothalamus detect leptin, and although the cellular and molecular mechanisms remain unclear, altered arcuate nucleus biology is hypothesized to contribute to selective leptin resistance. Male C57BL/6J mice were fed a high-fat diet (HFD) or chow from 8 to 18 weeks of age, as this paradigm models selective leptin resistance. Nuclei were then isolated from arcuate nucleus for single-nucleus RNA sequencing. HFD caused expected gains in adiposity and circulating leptin. Twenty-three unique cell-type clusters were identified, and Ingenuity Pathway Analysis was used to explore changes in gene expression patterns due to chronic HFD within each cluster. Notably, gene expression signatures related to leptin signaling exhibited suppression predominantly in neurons identified as the Agouti-related peptide (Agrp) subtype. Ingenuity Pathway Analysis results were also consistent with alterations in CREB (cAMP response element-binding protein) signaling in Agrp neurons after HFD, and reduced phosphorylated CREB was confirmed in arcuate nucleus after prolonged HFD by capillary electrophoresis-based Western blotting. These findings support the concept that prolonged HFD-induced obesity is associated with selective changes in Agrp neuron biology, possibly secondary to altered CREB signaling.


Subject(s)
Adiposity/physiology , Arcuate Nucleus of Hypothalamus/metabolism , Diet, High-Fat/adverse effects , Neurons/metabolism , Obesity/metabolism , Agouti-Related Protein/genetics , Agouti-Related Protein/metabolism , Animals , Cyclic AMP Response Element-Binding Protein/metabolism , Leptin/blood , Male , Mice , Obesity/etiology , Obesity/genetics , Phosphorylation , Sequence Analysis, RNA , Signal Transduction/physiology
18.
J Neuroendocrinol ; 31(8): e12752, 2019 08.
Article in English | MEDLINE | ID: mdl-31136029

ABSTRACT

Salt-loading (SL) impairs GABAA inhibition of arginine vasopressin (AVP) neurones in the supraoptic nucleus (SON) of the hypothalamus. Based on previous studies, we hypothesised that SL activates tyrosine receptor kinase B (TrkB), down-regulating the activity of K+ /Cl- co-transporter2 (KCC2) and up-regulating Na+ /K+ /Cl- co-transporter1 (NKCC1). These changes in chloride transport would result in increased [Cl- ]i in SON AVP neurones. The study combined virally-mediated chloride imaging with ClopHensorN with a single-cell western blot analysis. An adeno-associated virus with ClopHensorN and a vasopressin promoter (AAV2-0VP1-ClopHensorN) was bilaterally injected in the SON of adult male Sprague-Dawley rats that were either euhydrated (Eu) or salt-loaded (SL) for 7 days. Acutely dissociated SON neurones expressing ClopHensorN were tested for decreases or increases in [Cl- ]i in response to focal application of the GABAA agonist muscimol (100 µmol L-1 ). SON AVP neurones from Eu rats showed muscimol-induced chloride influx (P < 0.05;23/35). SON AVP neurones from SL rats either significantly increased chloride efflux (P < 0.05;27/39) or did not change chloride flux (12/39). The SON AVP neurones that responded to muscimol appeared to be viable and expressed KCC2 and ß-actin. Neurones that did not respond during chloride imaging did not show KCC2 and ß-actin protein expression. The KCC2 antagonist (VU0240551,10 µmol L-1 ) significantly blocked the chloride influx in cells from Eu rats but did not affect cells from SL rats. A NKCC1 antagonist (bumetanide,10 µmol L-1 ) significantly blocked the chloride efflux in cells from SL rats but had no effect on cells from Eu rats. Blocking NKCC1 using bumetanide had less of an effect on the muscimol-induced Cl- influx in Eu rat neurones compared to the KCC2 antagonist. The TrkB antagonist (AnA-12) (50 µmol L-1 ) and protein kinase inhibitor (K252a) (100 nmol L-1 ) each significantly blocked chloride efflux in SON AVP neurones from SL rats. Salt-loading increases [Cl- ]i in SON AVP neurones via a TrKB-KCC2-NKCC1-dependent mechanism in rats.


Subject(s)
Arginine Vasopressin/metabolism , Neurons/drug effects , Sodium Chloride/pharmacology , Supraoptic Nucleus/drug effects , Animals , Arginine Vasopressin/genetics , Biosensing Techniques , Dose-Response Relationship, Drug , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunohistochemistry/methods , Male , Neurons/cytology , Neurons/metabolism , Optical Imaging/methods , Promoter Regions, Genetic , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Supraoptic Nucleus/diagnostic imaging , Supraoptic Nucleus/metabolism
19.
J Neuroendocrinol ; 30(11): e12639, 2018 11.
Article in English | MEDLINE | ID: mdl-30129982

ABSTRACT

High salt loading (SL) is associated with inappropriate arginine vasopressin (AVP) release and increased mean arterial pressure. Previous work has shown that chronic high salt intake impairs baroreceptor inhibition of rat AVP neurones through brain-derived neurotrophic factor (BDNF) dependent activation of tyrosine receptor kinase B (TrkB) and down-regulation of K+/Cl- co-transporter KCC2. This mechanism diminishes the GABAA inhibition of AVP neurones in the supraoptic nucleus (SON) by increasing intracellular chloride. However, the source of BDNF leading to this ionic plasticity is unknown. In the present study, we used adeno-associated viral vectors with short hairpin RNA against BDNF to test whether SON is the source of BDNF contributing to increased AVP release and elevated mean arterial pressure in high salt loaded rats. Virally mediated BDNF knockdown (shBDNF) in the SON of salt loaded rats significantly blocked the increases in BDNF mRNA and AVP heterogeneous RNA expression. The observed increase in the activation of TrkB receptor during salt loading is consistent with previous studies. Western blot analysis of SON punches revealed that tyrosine phosphorylation of TrkB (pTrkBY515) was significantly decreased in salt shBDNF rats compared to the salt scrambled (SCR) rats. Injections of shBDNF in the SON also significantly prevented the increase in plasma AVP concentration associated with salt loading. However, the salt loading induced increase in mean arterial pressure was not decreased with BDNF knockdown in the SON. Average daily fluid intake and urine output were significantly elevated in both salt SCR and salt shBDNF rats compared to the euhydrated controls. Daily average urine sodium concentration was significantly higher in shBDNF injected salt rats than the other groups. These findings indicate that BDNF produced in the SON contributes to the increased AVP secretion during high salt loading but not with respect to the subsequent increase in mean arterial pressure.


Subject(s)
Arginine Vasopressin/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Neurons/metabolism , Sodium Chloride/administration & dosage , Supraoptic Nucleus/metabolism , Animals , Arterial Pressure , Eating , Gene Expression , Heart Rate , Male , Neurons/drug effects , Rats, Sprague-Dawley , Receptor, trkB/metabolism , Sodium/urine , Supraoptic Nucleus/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL