Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Mol Metab ; 45: 101161, 2021 03.
Article in English | MEDLINE | ID: mdl-33412345

ABSTRACT

OBJECTIVES: Heat-sensory neurons from the dorsal root ganglia (DRG) play a pivotal role in detecting the cutaneous temperature and transmission of external signals to the brain, ensuring the maintenance of thermoregulation. However, whether these thermoreceptor neurons contribute to adaptive thermogenesis remains elusive. It is also unknown whether these neurons play a role in obesity and energy metabolism. METHODS: We used genetic ablation of heat-sensing neurons expressing calcitonin gene-related peptide α (CGRPα) to assess whole-body energy expenditure, weight gain, glucose tolerance, and insulin sensitivity in normal chow and high-fat diet-fed mice. Exvivo lipolysis and transcriptional characterization were combined with adipose tissue-clearing methods to visualize and probe the role of sensory nerves in adipose tissue. Adaptive thermogenesis was explored using infrared imaging of intrascapular brown adipose tissue (iBAT), tail, and core temperature upon various stimuli including diet, external temperature, and the cooling agent icilin. RESULTS: In this report, we show that genetic ablation of heat-sensing CGRPα neurons promotes resistance to weight gain upon high-fat diet (HFD) feeding and increases energy expenditure in mice. Mechanistically, we found that loss of CGRPα-expressing sensory neurons was associated with reduced lipid deposition in adipose tissue, enhanced expression of fatty acid oxidation genes, higher exvivo lipolysis in primary white adipocytes, and increased mitochondrial respiration from iBAT. Remarkably, mice lacking CGRPα sensory neurons manifested increased tail cutaneous vasoconstriction at room temperature. This exacerbated cold perception was not associated with reduced core temperature, suggesting that heat production and heat conservation mechanisms were engaged. Specific denervation of CGRPα neurons in intrascapular BAT did not contribute to the increased metabolic rate observed upon global sensory denervation. CONCLUSIONS: Taken together, these findings highlight an important role of cutaneous thermoreceptors in regulating energy metabolism by triggering counter-regulatory responses involving energy dissipation processes including lipid fuel utilization and cutaneous vasodilation.


Subject(s)
Calcitonin Gene-Related Peptide/genetics , Calcitonin Gene-Related Peptide/metabolism , Obesity/metabolism , Sensory Receptor Cells/metabolism , Thermogenesis/genetics , Thermogenesis/physiology , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/pathology , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Animals , Body Temperature Regulation/genetics , Body Temperature Regulation/physiology , Cold Temperature , Diet, High-Fat/adverse effects , Energy Metabolism/genetics , Energy Metabolism/physiology , Female , Insulin Resistance , Lipolysis/genetics , Lipolysis/physiology , Male , Mice , Neurons
2.
Metabol Open ; 8: 100060, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33089134

ABSTRACT

OBJECTIVE: Calcitonin Gene-Related Peptide α (CGRPα) is a multifunctional neuropeptide found in the central and peripheral nervous system with cardiovascular, nociceptive, and gastrointestinal activities. CGRPα has been linked to obesity and insulin secretion but the role of this circulating peptide in energy metabolism remains unclear. Here, we thought to utilize a monoclonal antibody against circulating CGRPα to assess its ability to improve glucose homeostasis in mouse models of hyperglycemia and diabetes. METHODS: We examined the outcome of anti-CGRPα treatment in mouse models of diabetes and diet-induced obesity, using db/db mice, Streptozotocin (STZ) treatment to eliminate pancreatic islets, and high fat diet-fed mice. We also correlated these data with application of recombinant CGRPα peptide on cultured mature adipocytes to measure its impact on mitochondrial bioenergetics and fatty acid oxidation. Furthermore, we applied recombinant CGRPα to primary islets to measure glucose-stimulated insulin secretion (GSIS) and gene expression. RESULTS: BL6-db diabetic mice receiving anti-CGRPα treatment manifested weight loss, reduced adiposity, improved glucose tolerance, insulin sensitivity, GSIS and reduced pathology in adipose tissue and liver. Anti-CGRPα failed to modulate weight or glucose homeostasis in STZ-treated animals. High fat diet-fed mice showed reduced adiposity but no benefit on glucose homeostasis. Considering these findings, we postulated that CGRPα may have dual effects on adipocytes to promote lipid utilization while acting on pancreatic ß-cells to modulate insulin secretion. Analysis of CGRPα in the pancreas showed that the peptide localized to insulin-positive cells and perivascular nerves surrounding islets. Ex-vivo analysis of pancreatic islets determined that CGRPα blocked GSIS and reduced insulin-2 gene expression. Mechanistical analysis revealed that recombinant CGRPα was able to reduce glycolytic capacity as well as fatty acid oxidation in primary white adipocytes. CONCLUSIONS: These results establish a multifaceted role in energy metabolism for circulating CGRPα, with the ability to modulate thermogenic pathways in adipose tissue, as well as pancreatic ß-cell dependent insulin secretion. Reducing circulating CGRPα levels with monoclonal therapy presents therapeutic potential for type 2 diabetes as shown in BL6-db/db mice but has reduced potential for models of hyperglycemia resulting from loss of ß-cells (STZ treatment).

3.
Physiol Rep ; 8(3): e14364, 2020 02.
Article in English | MEDLINE | ID: mdl-32026607

ABSTRACT

Alteration in renin-angiotensin system (RAS) has been implicated in the pathophysiology of diabetic kidney disease (DKD). The deleterious actions of angiotensin II (Ang II) could be antagonized by the formation of Ang-(1-7), generated by the actions of angiotensin-converting enzyme 2 (ACE2) and neprilysin (NEP). NEP degrades several peptides, including natriuretic peptides, bradykinin, amyloid beta, and Ang I. Although combination of Ang II receptor and NEP inhibitor treatment benefits patients with heart failure, the role of NEP in renal pathophysiology is a matter of active research. NEP pathway is a potent enzyme in Ang I to Ang-(1-7) conversion in the kidney of ACE2-deficient mice, suggesting a renoprotective role of NEP. The aim of the study is to test the hypothesis that chronic hyperglycemia downregulates renal NEP protein expression and activity in db/db diabetic mice and treatment with rosiglitazone normalizes hyperglycemia, renal NEP expression, and attenuates albuminuria. Mice received rosiglitazone (20 mg kg-1  day-1 ) for 10 weeks. Western blot analysis, immunohistochemistry, and enzyme activity revealed a significant decrease in renal and urinary NEP expression and activity in 16-wk db/db mice compared with lean control (p < .0001). Rosiglitazone also attenuated albuminuria and increased renal and urinary NEP expressions (p < .0001). In conclusion, data support the hypothesis that diabetes decreases intrarenal NEP, which could have a pivotal role in the pathogenesis of DKD. Urinary NEP may be used as an index of intrarenal NEP status. The renoprotective effects of rosiglitazone could be mediated by upregulation of renal NEP expression and activity in db/db diabetic mice.


Subject(s)
Diabetic Nephropathies/metabolism , Hyperglycemia/metabolism , Hypoglycemic Agents/therapeutic use , Neprilysin/metabolism , Rosiglitazone/therapeutic use , Animals , Diabetic Nephropathies/drug therapy , Down-Regulation , Hyperglycemia/drug therapy , Hypoglycemic Agents/pharmacology , Kidney/drug effects , Kidney/metabolism , Male , Mice , Neprilysin/urine , Rosiglitazone/pharmacology
4.
J Endocr Soc ; 1(4): 370-384, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-29082356

ABSTRACT

Angiotensin-converting enzyme 2 (ACE2) has protective effects on a wide range of morbidities associated with elevated angiotensin-II signaling. Most tissues, including pancreatic islets, express ACE2 mainly from the proximal promoter region. We previously found that hepatocyte nuclear factors 1α and 1ß stimulate ACE2 expression from three highly conserved hepatocyte nuclear factor 1 binding motifs in the proximal promoter region. We hypothesized that other highly conserved motifs would also affect ACE2 expression. By systematic mutation of conserved elements, we identified five regions affecting ACE2 expression, of which two regions bound transcriptional activators. One of these is a functional FOXA binding motif. We further identified the main protein binding the FOXA motif in 832/13 insulinoma cells as well as in mouse pancreatic islets as FOXA2.

5.
Am J Physiol Regul Integr Comp Physiol ; 311(6): R1223-R1233, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27806985

ABSTRACT

While restoration of ACE2 activity in the pancreas leads to improvement of glycemia in experimental models of Type 2 diabetes, global deficiency in ACE2 disrupts ß-cell function and impairs glucose tolerance in mice, demonstrating the physiological role of ACE2 in glucose homeostasis. Although the contribution of pancreatic ACE2 to glucose regulation has been demonstrated in genetic models of diabetes and in models with overexpression of the renin-angiotensin system (RAS), it is unclear whether islet ACE2 is involved in glycemic control in common models of human Type 2 diabetes. To determine whether diet-induced diabetes deregulates glucose homeostasis via reduction of ACE2 in the pancreatic islets, wild-type (WT) and ACE2 knockout (KO) male mice were fed a high-fat diet (HFD) for 16 wk. ACE2 KO mice were more susceptible than WT mice to HFD-mediated glycemic dysregulation. Islet ACE2 activity and expression of various genes, including ANG II type 1a receptor (mAT1aR) were then assessed. Surprisingly, we observed no change in islet ACE2 activity and expression despite local RAS overactivity, indicated by an upregulation of mAT1aR expression. Despite a predominant expression in islet α-cells, further investigation highlighted a minor role for ACE2 on glucagon expression. Further, pancreatic ACE2 gene therapy improved glycemia in HFD-fed WT mice, leading to enhanced glucose-stimulated insulin secretion, reduced pancreatic ANG II levels, fibrosis, and ADAM17 activity. Altogether, our study demonstrates that HFD feeding increases RAS activity and mediates glycemic dysregulation likely through loss of ACE2 present outside the islets but independently of changes in islet ACE2.


Subject(s)
Diet, High-Fat/adverse effects , Glucose Metabolism Disorders/etiology , Glucose Metabolism Disorders/metabolism , Glucose/metabolism , Islets of Langerhans/metabolism , Peptidyl-Dipeptidase A/metabolism , Angiotensin-Converting Enzyme 2 , Animals , Dietary Fats/adverse effects , Insulin/metabolism , Insulin Resistance , Insulin Secretion , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
6.
Endocrinology ; 156(12): 4411-25, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26441236

ABSTRACT

Angiotensin-converting enzyme 2 (ACE2) gene therapy aimed at counteracting pancreatic ACE2 depletion improves glucose regulation in two diabetic mouse models: db/db mice and angiotensin II-infused mice. A disintegrin and metalloproteinase 17 (ADAM17) can cause shedding of ACE2 from the cell membrane. The aim of our studies was to determine whether ADAM17 depletes ACE2 levels in pancreatic islets and ß-cells. Dynamics of ADAM17-mediated ACE2 shedding were investigated in 832/13 insulinoma cells. Within a wide range of ACE2 expression levels, including the level observed in mouse pancreatic islets, overexpression of ADAM17 increases shed ACE2 and decreases cellular ACE2 levels. We provide a mathematical description of shed and cellular ACE2 activities as a function of the ADAM17 activity. The effect of ADAM17 on the cellular ACE2 content was relatively modest with an absolute control strength value less than 0.25 and approaching 0 at low ADAM17 activities. Although we found that ADAM17 and ACE2 are both expressed in pancreatic islets, the ß-cell is not the major cell type expressing ACE2 in islets. During diabetes progression in 8-, 12-, and 15-week-old db/db mice, ACE2 mRNA and ACE2 activity levels in pancreatic islets were not decreased over time nor significantly decreased compared with nondiabetic db/m mice. Levels of ADAM17 mRNA and ADAM17 activity were also not significantly changed. Inhibiting basal ADAM17 activity in mouse islets failed to affect ACE2 levels. We conclude that whereas ADAM17 has the ability to shed ACE2, ADAM17 does not deplete ACE2 from pancreatic islets in diabetic db/db mice.


Subject(s)
ADAM Proteins/genetics , Cell Membrane/metabolism , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus/genetics , Insulin-Secreting Cells/metabolism , Peptidyl-Dipeptidase A/genetics , RNA, Messenger/metabolism , ADAM Proteins/metabolism , ADAM17 Protein , Angiotensin II , Angiotensin-Converting Enzyme 2 , Animals , Cell Line, Tumor , Diabetes Mellitus/metabolism , Diabetes Mellitus, Experimental/metabolism , Disease Progression , Insulinoma/metabolism , Islets of Langerhans/metabolism , Male , Mice , Pancreatic Neoplasms/metabolism , Peptidyl-Dipeptidase A/metabolism , Rats
7.
Am J Physiol Cell Physiol ; 308(9): C767-77, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25740155

ABSTRACT

The renin angiotensin system (RAS) plays a vital role in the regulation of the cardiovascular and renal functions. COS7 is a robust and easily transfectable cell line derived from the kidney of the African green monkey, Cercopithecus aethiops. The aims of this study were to 1) demonstrate the presence of an endogenous and functional RAS in COS7, and 2) investigate the role of a disintegrin and metalloproteinase-17 (ADAM17) in the ectodomain shedding of angiotensin converting enzyme-2 (ACE2). Reverse transcription coupled to gene-specific polymerase chain reaction demonstrated expression of ACE, ACE2, angiotensin II type 1 receptor (AT1R), and renin at the transcript levels in total RNA cell extracts. Western blot and immunohistochemistry identified ACE (60 kDa), ACE2 (75 kDa), AT1R (43 kDa), renin (41 kDa), and ADAM17 (130 kDa) in COS7. At the functional level, a sensitive and selective mass spectrometric approach detected endogenous renin, ACE, and ACE2 activities. ANG-(1-7) formation (m/z 899) from the natural substrate ANG II (m/z 1,046) was detected in lysates and media. COS7 cells stably expressing shRNA constructs directed against endogenous ADAM17 showed reduced ACE2 shedding into the media. This is the first study demonstrating endogenous expression of the RAS and ADAM17 in the widely used COS7 cell line and its utility to study ectodomain shedding of ACE2 mediated by ADAM17 in vitro. The transfectable nature of this cell line makes it an attractive cell model for studying the molecular, functional, and pharmacological properties of the renal RAS.


Subject(s)
ADAM Proteins/metabolism , Peptidyl-Dipeptidase A/metabolism , Renin-Angiotensin System , ADAM Proteins/genetics , ADAM17 Protein , Angiotensin I/metabolism , Angiotensin II/genetics , Angiotensin II/metabolism , Angiotensin-Converting Enzyme 2 , Animals , Base Sequence , COS Cells , Chlorocebus aethiops , Molecular Sequence Data , Peptide Fragments/metabolism , Peptidyl-Dipeptidase A/genetics , RNA Interference , RNA, Messenger/metabolism , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism , Renin/genetics , Renin/metabolism , Renin-Angiotensin System/genetics , Transcription, Genetic , Transfection
8.
Muscles Ligaments Tendons J ; 4(3): 280-4, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25489543

ABSTRACT

Foot ulcers are a severe complication of diabetic patients resulting from nerve and tendon pathologic alterations. In diabetic patients the tendons are thicker, shorter and have increased stiffness. We examined C57BL/KsJ (BKS.Cg-Dock7(m) +/+ Lepr (db) /J) (db/db) mice tendons to determine whether they are an animal model for human diabetic tendon changes. We hypothesized that the Achilles tendons of db/db diabetic mice would be thicker, stiffer, fail at lower loads and stresses, and have degenerative changes compared to control mice. Biomechanical and histologic analyses of the Achilles tendons of 16 week old db/db and control male mice were performed. There was a significant increase in tendon diameter and significant decreases in maximum load, tensile stress, stiffness and elastic modulus in tendons from diabetic mice compared to controls. Mild degenerative and neutrophil infiltration was observed near the tendon insertions on the calcaneous in 25% of db/db mice. In summary, hyper-glycemia and obesity lead to severe changes in db/db mice will be a useful model to examine mechanisms for tendon alterations.

9.
IUBMB Life ; 65(9): 731-8, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23893738

ABSTRACT

In spite of the novel antidiabetic drugs available on the market, type 2 diabetes mellitus (T2DM) affects nearly 25 million people in the USA and causes about 5% of all deaths globally each year. Given the rate and proportion by which T2DM is affecting human beings, it is indispensable to identify new therapeutic targets that can control the disease. Recent preclinical and clinical studies suggest that attenuating the activity of the renin-angiotensin system (RAS) could improve glycemia in diabetic patients. Angiotensin-converting enzyme 2 (ACE2) counteracts RAS overactivity by degrading angiotensin-II (Ang-II), a vasoconstrictor, to Ang-(1-7) which is a vasodilator. A decrease in ACE2 and an increase in A disintegrin and metalloproteinase (ADAM17)-mediated shedding activity have been observed with the progression of T2DM, suggesting the importance of this mechanism in the disease. Indeed, restoration of ACE2 improves glycemia in db/db and Ang-II-infused mice. The beneficial effects of ACE2 can be attributed to reduced oxidative stress and ADAM17 expression in the islets of Langerhans in addition to the improvement of blood flow to the ß-cells. The advantage of ACE2 over other RAS blockers is that ACE2 not only counteracts the negative effects of Ang-II but also increases Ang-(1-7)/Mas receptor (MasR) [a receptor through which Ang-(1-7) produces its actions] signaling in the cells. Increased Ang-(1-7)/MasR signaling has been reported to improve insulin sensitivity and glycemia in diabetic animals. Altogether, ACE2/Ang-(1-7)/MasR axis of the RAS appears to be protective in T2DM and strategies to restore ACE2 levels in the disease seem to be a promising therapy for Ang-II-mediated T2DM.


Subject(s)
Diabetes Mellitus, Type 2/blood , Peptidyl-Dipeptidase A/physiology , Angiotensin-Converting Enzyme 2 , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Animals , Blood Glucose , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/enzymology , Homeostasis , Humans , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Proto-Oncogene Mas , Renin-Angiotensin System
10.
PLoS One ; 8(4): e62833, 2013.
Article in English | MEDLINE | ID: mdl-23646149

ABSTRACT

Alterations within the renal renin angiotensin system play a pivotal role in the development and progression of cardiovascular and renal disease. Angiotensin converting enzyme 2 (ACE2) is highly expressed in renal tubules and has been shown to be renoprotective in diabetes. The protease, a disintegrin and metalloprotease (ADAM) 17, is involved in the ectodomain shedding of several transmembrane proteins including ACE2. Renal ACE2 and ADAM17 were significantly increased in db/db mice compared to controls. We investigated the effect of the insulin sensitizer, rosiglitazone, on albuminuria, renal ADAM17 protein expression and ACE2 shedding in db/db diabetic mice. Rosiglitazone treatment of db/db mice normalized hyperglycemia, attenuated renal injury and decreased urinary ACE2 and renal ADAM17 protein expression. Urinary excreted ACE2 is enzymatically active. Western blot analysis of urinary ACE2 demonstrated two prominent immunoreactive bands at approximately 70 & 90 kDa. The predominant immunoreactive band is approximately 20 kDa shorter than the one demonstrated for kidney lysate, indicating possible ectodomain shedding of active renal ACE2 in the urine. Therefore, it is tempting to speculate that renoprotection of rosiglitazone could be partially mediated via downregulation of renal ADAM17 and ACE2 shedding. In addition, there was a positive correlation between blood glucose, urinary albumin, plasma glucagon, and triglyceride levels with urinary ACE2 excretion. In conclusion, urinary ACE2 could be used as a sensitive biomarker of diabetic nephropathy and for monitoring the effectiveness of renoprotective medication.


Subject(s)
Albuminuria/urine , Diabetes Mellitus, Type 2/urine , Hypoglycemic Agents/pharmacology , Peptidyl-Dipeptidase A/urine , Thiazolidinediones/pharmacology , ADAM Proteins/metabolism , ADAM17 Protein , Angiotensin-Converting Enzyme 2 , Animals , Blood Glucose/drug effects , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Disease Models, Animal , Glucagon/blood , Glucose Tolerance Test , Glycosuria , Hyperglycemia/drug therapy , Hypoglycemic Agents/administration & dosage , Kidney Diseases/drug therapy , Kidney Diseases/metabolism , Kidney Diseases/pathology , Male , Membrane Proteins/metabolism , Mice , Peptidyl-Dipeptidase A/metabolism , Rosiglitazone , Thiazolidinediones/administration & dosage , Tissue Inhibitor of Metalloproteinase-3/metabolism , Triglycerides/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...