Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Life Sci ; 283: 119841, 2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34298036

ABSTRACT

Sympathetic vasomotor overactivity is a major feature leading to the cardiovascular dysfunction related to obesity. Considering that the retroperitoneal white adipose tissue (rWAT) is an important fat visceral depot and receives intense sympathetic and afferent innervations, the present study aimed to evaluate the effects evoked by bilateral rWAT denervation in obese rats. Male Wistar rats were fed with HFD for 8 consecutive weeks and rWAT denervation was performed at the 6th week. Arterial pressure, splanchnic and renal sympathetic vasomotor nerve activities were assessed and inflammation and the components of the renin -angiotensin system were evaluated in different white adipose tissue depots. HFD animals presented higher serum levels of leptin and glucose, an increase in arterial pressure and splanchnic sympathetic nerve activity; rWAT denervation, normalized these parameters. Pro-inflammatory cytokines levels were significantly increased, as well as RAAS gene expression in WAT of HFD animals; rWAT denervation significantly attenuated these changes. In conclusion, HFD promotes vasomotor sympathetic overactivation and inflammation with repercussions on the cardiovascular system. In conclusion, the neural communication between WAT and the brain is fundamental to trigger sympathetic vasomotor activation and this pathway is a possible new therapeutic target to treat obesity-associated cardiovascular dysfunction.


Subject(s)
Cardiovascular Diseases , Denervation , Diet, High-Fat/adverse effects , Intra-Abdominal Fat , Obesity , Splanchnic Nerves , Animals , Blood Pressure , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/physiopathology , Cardiovascular Diseases/therapy , Cardiovascular System/metabolism , Cardiovascular System/physiopathology , Intra-Abdominal Fat/innervation , Intra-Abdominal Fat/metabolism , Intra-Abdominal Fat/physiopathology , Male , Obesity/chemically induced , Obesity/metabolism , Obesity/physiopathology , Obesity/therapy , Rats , Rats, Wistar , Renin-Angiotensin System , Splanchnic Nerves/metabolism , Splanchnic Nerves/pathology , Splanchnic Nerves/physiopathology
2.
Physiol Behav ; 190: 28-33, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29447836

ABSTRACT

White adipose tissue (WAT) and brown adipose tissue (BAT) have sympathetic nervous system (SNS) and sensory innervations. Previous studies from our laboratory revealed central neuroanatomical evidence of WAT sensory and BAT SNS crosstalk with double labeling of inguinal WAT (IWAT) sensory and interscapular BAT (IBAT) SNS neurons. We previously demonstrated that WAT lipolysis increases IBAT temperature, but this effect is absent when IWAT afferents are surgically denervated, which severs both sensory and SNS nerves. It is possible that WAT sensory feedback can regulate SNS drive to itself and other WAT and BAT depots, and thus contribute to the existence of differential SNS outflow to fat during different energy challenges. Here we selectively denervated IWAT sensory nerves in Siberian hamsters using capsaicin and measured norepinephrine turnover (NETO) i.e., SNS drive to WAT and BAT depots, IBAT uncoupling protein 1 (UCP1) expression, body mass, fat mass, blood glucose, and food consumed after a 24-h cold exposure. IWAT sensory denervation decreased both IWAT and IBAT NETO and IBAT UCP1 expression. IWAT sensory denervation, however, increased mesenteric WAT (MWAT) NETO after the 24-h cold exposure and did not modify epididymal WAT (EWAT) and retroperitoneal WAT (RWAT) NETO compared with respective controls. Body mass, fat mass, blood glucose, and food consumed were unchanged across groups. RWAT and EWAT mass decreased in capsaicin-injected hamsters, but did not in the vehicle hamsters. These results functionally demonstrate the existence of IWAT sensory and IBAT SNS crosstalk and that a disruption in this sensory-SNS feedback mechanism modifies SNS drive to IWAT, IBAT, and MWAT, but not EWAT and RWAT.


Subject(s)
Adipose Tissue, Brown/innervation , Adipose Tissue, White/innervation , Denervation , Intra-Abdominal Fat/innervation , Sympathetic Nervous System/physiology , Animals , Blood Glucose/metabolism , Body Mass Index , Capsaicin , Cricetinae , Eating/physiology , Male , Norepinephrine/metabolism , Peripheral Nerves/drug effects , Phodopus , Uncoupling Protein 1/biosynthesis
3.
Cell Metab ; 27(1): 226-236.e3, 2018 01 09.
Article in English | MEDLINE | ID: mdl-29320703

ABSTRACT

While the cell-intrinsic pathways governing beige adipocyte development and phenotype have been increasingly delineated, comparatively little is known about how beige adipocytes interact with other cell types in fat. Here, we introduce a whole-tissue clearing method for adipose that permits immunolabeling and three-dimensional profiling of structures including thermogenic adipocytes and sympathetic innervation. We found that tissue architecture and sympathetic innervation differ significantly between subcutaneous and visceral depots. Subcutaneous fat demonstrates prominent regional variation in beige fat biogenesis with localization of UCP1+ beige adipocytes to areas with dense sympathetic neurites. We present evidence that the density of sympathetic projections is dependent on PRDM16 in adipocytes, providing another potential mechanism underlying the metabolic benefits mediated by PRDM16. This powerful imaging tool highlights the interaction of tissue components during beige fat biogenesis and reveals a previously undescribed mode of regulation of the sympathetic nervous system by adipocytes.


Subject(s)
Adipose Tissue, Beige/anatomy & histology , Adipose Tissue, Beige/metabolism , DNA-Binding Proteins/metabolism , Imaging, Three-Dimensional , Neurites/metabolism , Sympathetic Nervous System/metabolism , Transcription Factors/metabolism , Adipocytes/metabolism , Adipose Tissue, Beige/innervation , Animals , Intra-Abdominal Fat/innervation , Intra-Abdominal Fat/metabolism , Mice, Inbred C57BL , Mice, Knockout , Subcutaneous Fat/innervation , Subcutaneous Fat/metabolism
4.
Nutr Diabetes ; 7(4): e260, 2017 04 10.
Article in English | MEDLINE | ID: mdl-28394360

ABSTRACT

The sympathetic nervous system (SNS) regulates energy homeostasis in part by governing fatty acid liberation from adipose tissue. We first examined whether SNS activity toward discrete adipose depots changes in response to a weight loss diet in mice. We found that SNS activity toward each adipose depot is unique in timing, pattern of activation, and habituation with the most dramatic contrast between visceral and subcutaneous adipose depots. Sympathetic drive toward visceral epididymal adipose is more than doubled early in weight loss and then suppressed later in the diet when weight loss plateaued. Coincident with the decline in SNS activity toward visceral adipose is an increase in activity toward subcutaneous depots indicating a switch in lipolytic sources. In response to calorie restriction, SNS activity toward retroperitoneal and brown adipose depots is unaffected. Finally, pharmacological blockage of sympathetic activity on adipose tissue using the ß3-adrenergic receptor antagonist, SR59230a, suppressed loss of visceral adipose mass in response to diet. These findings indicate that SNS activity toward discrete adipose depots is dynamic and potentially hierarchical. This pattern of sympathetic activation is required for energy liberation and loss of adipose tissue in response to calorie-restricted diet.


Subject(s)
Caloric Restriction , Diet, Reducing , Energy Intake , Intra-Abdominal Fat/metabolism , Norepinephrine/metabolism , Obesity/metabolism , Sympathetic Nervous System/physiology , Adipose Tissue, Brown/innervation , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/innervation , Adipose Tissue, White/metabolism , Adiposity , Adrenergic beta-3 Receptor Antagonists/pharmacology , Animals , Body Weight , Energy Metabolism , Epididymis/innervation , Epididymis/metabolism , Intra-Abdominal Fat/innervation , Lipolysis , Male , Mice, Inbred C57BL , Obesity/diet therapy , Peritoneum , Propanolamines/pharmacology , Subcutaneous Fat/innervation , Subcutaneous Fat/metabolism , Weight Loss
5.
PLoS One ; 12(3): e0173934, 2017.
Article in English | MEDLINE | ID: mdl-28346471

ABSTRACT

BACKGROUND: Interruption of sympathetic innervation to the liver and visceral adipose tissue (VAT) in animal models has been reported to reduce VAT lipolysis and hepatic secretion of very low density lipoprotein (VLDL) and concentrations of triglyceride-rich lipoprotein particles. Whether functional impairment of sympathetic nervous system (SNS) innervation to tissues of the abdominal cavity reduce circulating concentrations of triglyceride (TG) and VLDL particles (VLDL-P) was tested in men with spinal cord injury (SCI). METHODS: One hundred-three non-ambulatory men with SCI [55 subjects with neurologic injury at or proximal to the 4th thoracic vertebrae (↑T4); 48 subjects with SCI at or distal to the 5th thoracic vertebrae (↓T5)] and 53 able-bodied (AB) subjects were studied. Fasting blood samples were obtained for determination of TG, VLDL-P concentration by NMR spectroscopy, serum glucose by autoanalyzer, and plasma insulin by radioimmunoassay. VAT volume was determined by dual energy x-ray absorptiometry imaging with calculation by a validated proprietary software package. RESULTS: Significant group main effects for TG and VLDL-P were present; post-hoc tests revealed that serum TG concentrations were significantly higher in ↓T5 group compared to AB and ↑T4 groups [150±9 vs. 101±8 (p<0.01) and 112±8 mg/dl (p<0.05), respectively]. VLDL-P concentration was significantly elevated in ↓T5 group compared to AB and ↑T4 groups [74±4 vs. 58±4 (p<0.05) and 55±4 µmol/l (p<0.05)]. VAT volume was significantly higher in both SCI groups than in the AB group, and HOMA-IR was higher and approached significance in the SCI groups compared to the AB group. A linear relationship between triglyceride rich lipoproteins (i.e., TG or Large VLDL-P) and VAT volume or HOMA-IR was significant only in the ↓T5 group. CONCLUSIONS: Despite a similar VAT volume and insulin resistance in both SCI groups, the ↓T5 group had significantly higher serum TG and VLDL-P values than that observed in the ↑T4 and the AB control groups. Thus, level of injury is an important determinate of the concentration of circulating triglyceride rich lipoproteins, which may play a role in the genesis of cardiometabolic dysfunction.


Subject(s)
Intra-Abdominal Fat/innervation , Lipoproteins, VLDL/blood , Lipoproteins/blood , Liver/innervation , Spinal Cord Injuries/blood , Spinal Cord Injuries/physiopathology , Sympathetic Nervous System/physiopathology , Triglycerides/blood , Adult , Animals , Cohort Studies , Humans , Intra-Abdominal Fat/physiopathology , Liver/physiopathology , Male , Middle Aged
6.
Can J Physiol Pharmacol ; 94(5): 534-41, 2016 May.
Article in English | MEDLINE | ID: mdl-26963333

ABSTRACT

Increasing evidence indicates a link between sympathetic nervous system activation and obesity, but the underlying mechanisms remain elusive. The adipose afferent reflex (AAR) is a sympathoexcitatory reflex that is activated by afferent neurotransmission from the white adipose tissue (WAT). This study aimed to investigate whether the hypothalamic paraventricular nucleus (PVH) is an important component of the central neurocircuitry of the AAR. In anesthetized rats, the discharge activity of individual PVH neurons was recorded in vivo. Activation of WAT afferents was initiated by capsaicin injection, and the AAR was evaluated by monitoring renal sympathetic nerve activity (RSNA) and mean arterial pressure (MAP) responses. The responses of PVH neurons to activation of WAT afferents were evaluated by c-fos immunoreactivity and the discharge activity of individual PVH neurons, which was recorded using extracellular single-unit recording. After activation of WAT afferents, both individual PVH neuron discharge activity and c-fos immunoreactivity increased. Bilateral selective lesions of the neurons in the PVH with kainic acid abolished the AAR. These results indicate that PVH is an important component of the central neurocircuitry of the AAR.


Subject(s)
Adipose Tissue, White/innervation , Models, Neurological , Neurons, Afferent/physiology , Paraventricular Hypothalamic Nucleus/physiology , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Animals , Biomarkers/metabolism , Blood Pressure/drug effects , Capsaicin/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Inguinal Canal , Intra-Abdominal Fat/drug effects , Intra-Abdominal Fat/innervation , Intra-Abdominal Fat/metabolism , Kainic Acid/pharmacology , Male , Nerve Tissue Proteins/metabolism , Neurons, Afferent/drug effects , Paraventricular Hypothalamic Nucleus/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Rats, Sprague-Dawley , Retroperitoneal Space , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/metabolism , Sympathetic Nervous System/physiology , Synaptic Transmission/drug effects
7.
Chem Biol Interact ; 229: 1-8, 2015 Mar 05.
Article in English | MEDLINE | ID: mdl-25637688

ABSTRACT

Key features of diet-induced obesity are visceral fat deposition, macrophage infiltration and inflammation that can lead to metabolic disorders. This study examined the effects of bardoxolone methyl (BARD) in preventing obesity and inflammation in the visceral fat of mice fed high-fat diet. Male C57BL/6J mice were fed a high-fat diet (HFD), a low-fat diet (LFD, i.e., lab chow diet) or a high-fat diet supplemented with BARD (HFD/BARD) for 21weeks. BARD at a dosage of 10mg/kg body weight was administered orally in drinking water. Histology, immunohistochemistry and Western blot were used for the analysis of epididymal adipose tissue. Morphological results demonstrated that HFD fed mice treated with BARD had smaller adipocytes and fewer macrophages present in epididymal adipose tissue than the HFD group. Furthermore, BARD administration reduced the inflammatory profile in this tissue by increasing the expression of nuclear factor of kappa-light-polypeptide gene enhancer in B-cells inhibitor, alpha (IκB-α) protein and decreasing the protein expression of tumour necrosis factor alpha (TNF-α). BARD also prevented oxidative stress reflected by a reduction in stress activated proteins, including signal transducer and activator of transcription 3 (STAT3), protein kinase B (Akt), extracellular-signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK). BARD administration activated the sympathetic nervous system in epididymal adipose tissue assessed by the increased synthesis of tyrosine hydroxylase (TH) and uncoupling protein 2 (UCP2). The expression of inflammatory and sympathetic nervous system proteins in BARD mice fed a HFD was equivalent to that of the LFD control mice, indicating the anti-inflammatory and anti-obesity properties of this drug. In conclusion, the oral administration of BARD in HFD mice prevented fat deposition, inflammation and oxidative stress, and improved sympathetic activity in visceral fat. This study suggests a potential therapeutic role of BARD in preventing the development of obesity.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Diet, High-Fat/adverse effects , Inflammation/prevention & control , Intra-Abdominal Fat/drug effects , Obesity/prevention & control , Oleanolic Acid/analogs & derivatives , Adipocytes/cytology , Adipocytes/drug effects , Animals , Inflammation/immunology , Inflammation/metabolism , Intra-Abdominal Fat/immunology , Intra-Abdominal Fat/innervation , Intra-Abdominal Fat/metabolism , Macrophages/cytology , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Obesity/immunology , Obesity/metabolism , Oleanolic Acid/therapeutic use , Oxidative Stress/drug effects , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/immunology
8.
Obesity (Silver Spring) ; 23(1): 105-11, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25322680

ABSTRACT

OBJECTIVES: To determine whether a selective increase of visceral adipose tissue content will result in insulin resistance. METHODS: Sympathetic denervation of the omental fat was performed under general inhalant anesthesia by injecting 6-hydroxydopamine in the omental fat of lean mongrel dogs (n = 11). In the conscious animal, whole-body insulin sensitivity was assessed by the minimal model (SI ) and the euglycemic hyperinsulinemic clamp (SICLAMP ). Changes in abdominal fat were monitored by magnetic resonance. All assessments were determined before (Wk0) and 2 weeks (Wk2) after denervation. Data are medians (upper and lower interquartile). RESULTS: Denervation of omental fat resulted in increased percentage (and content) of visceral fat [Wk0: 10.2% (8.5-11.4); Wk2: 12.4% (10.4-13.6); P < 0.01]. Abdominal subcutaneous fat remained unchanged. However, no changes were found in SI [Wk0: 4.7 (mU/l)(-1) min(-1) (3.1-8.8); Wk2: 5.3 (mU/l)(-1) min(-1) (4.5-7.2); P = 0.59] or SICLAMP [Wk0: 42.0 × 10(-4) dl kg(-1) min(-1) (mU/l)(-1) (41.0-51.0); Wk2: 40.0 × 10(-4) dl kg(-1) min(-1) (mU/l) (-1) (34.0-52.0); P = 0.67]. CONCLUSIONS: Despite a selective increase in visceral adiposity in dogs, insulin sensitivity in vivo did not change, which argues against the concept that accumulation of visceral adipose tissue contributes to insulin resistance.


Subject(s)
Insulin Resistance , Intra-Abdominal Fat/anatomy & histology , Intra-Abdominal Fat/metabolism , Animals , Body Composition , Body Weight , Dogs , Glucose Clamp Technique , Intra-Abdominal Fat/innervation , Magnetic Resonance Imaging , Male , Models, Animal , Omentum/innervation , Organ Size , Subcutaneous Fat, Abdominal/anatomy & histology , Subcutaneous Fat, Abdominal/innervation , Subcutaneous Fat, Abdominal/metabolism , Sympathectomy, Chemical/veterinary
9.
Am J Physiol Regul Integr Comp Physiol ; 306(6): R375-86, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24452544

ABSTRACT

There is a link between visceral white adipose tissue (WAT) and the metabolic syndrome in humans, with health improvements produced with small visceral WAT reduction. By contrast, subcutaneous WAT provides a site for lipid storage that is rather innocuous relative to ectopic lipid storage in muscle or liver. The sympathetic nervous system (SNS) is the principal initiator for lipolysis in WAT by mammals. Nothing is known, however, about the central origins of the SNS circuitry innervating the only true visceral WAT in rodents, mesenteric WAT (MWAT), which drains into the hepatic portal vein. We tested whether the central sympathetic circuits to subcutaneous [inguinal WAT (IWAT)] and visceral WAT (MWAT) are separate or shared and whether they possess differential sympathetic drives with food deprivation in Siberian hamsters. Using two isogenic strains of pseudorabies virus, a retrograde transneuronal viral tract tracer within the same hamsters, we found some overlap (∼20-55% doubly infected neurons) between the two circuitries across the neural axis with lesser overlap proximal to the depots (spinal cord and sympathetic chain) and with more neurons involved in the innervation of IWAT than MWAT in some brain regions. Food deprivation triggered a greater sympathetic drive to subcutaneous (IWAT) than visceral (MWAT) depots. Collectively, we demonstrated both shared and separate populations of brain, spinal cord, and sympathetic chain neurons ultimately project to a subcutaneous WAT depot (IWAT) and the only visceral WAT depot in rodents (MWAT). In addition, the lipolytic stimulus of food deprivation only increased SNS drive to subcutaneous fat (IWAT).


Subject(s)
Adipose Tissue, White/innervation , Central Nervous System/cytology , Food Deprivation/physiology , Ganglia, Sympathetic/cytology , Intra-Abdominal Fat/innervation , Subcutaneous Fat/innervation , Adipose Tissue, White/metabolism , Adrenergic Fibers/physiology , Animals , Central Nervous System/metabolism , Cricetinae , Ganglia, Sympathetic/metabolism , Herpesvirus 1, Suid , Intra-Abdominal Fat/metabolism , Lipolysis/physiology , Male , Neuronal Tract-Tracers , Phodopus , Subcutaneous Fat/metabolism
10.
Hepatogastroenterology ; 59(116): 1299-301, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22580681

ABSTRACT

BACKGROUND/AIMS: Although preservation of the vaguas nerve is recommended in surgery for earlystage gastric cancer, the physiological effect of vagotomy on the postoperative course has not been well documented. We assessed the effect of vagotomy on the change in fat volume after gastrectomy. METHODOLOGY: Subcutaneous fat area (SFA) and visceral fat area (VFA) were separately measured in computed tomographic images taken before and more than 6 months after surgery, using Fat Scan software. The ratios of postoperative/ preoperative values of these two fat areas as well as body weight were calculated in 45 patients who underwent DG with (n=24) or without (n=21) vagotomy. RESULTS: Vagotomy did not affect the change in body weight (91.3±1.7% vs. 92.1±1.7%). In patients with vagotomy, VFA was reduced to 59.0±5.1%, which was significantly greater than the reduction in SFA (74.3±8.7%, p=0.042). In contrast, the reduction ratios of VFA and SFA were equal in vagus nerve-preserved patients (78.4±6.7% vs. 78.2±6.9%, p=0.97). CONCLUSIONS: The vagus nerve may have a function to locally regulate the intra-abdominal fat volume and preservation of the vagus nerve results in the maintenance of visceral fat after DG.


Subject(s)
Gastrectomy/methods , Intra-Abdominal Fat/physiology , Vagotomy , Aged , Body Weight , Female , Humans , Intra-Abdominal Fat/innervation , Male , Middle Aged , Retrospective Studies , Subcutaneous Fat/innervation , Subcutaneous Fat/physiology , Vagus Nerve/physiology
11.
J Pediatr Surg ; 46(12): 2358-62, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22152882

ABSTRACT

BACKGROUND/PURPOSE: Inguinoscrotal testicular descent is controlled by androgens between embryonic days E16-19, but androgen receptor (AR) and estrogen receptor (ER) locations are unknown. We aimed to find AR, ERα, and ERß in the gubernaculum and inguinal fat pad (IFP) in normal rats and after flutamide treatment. METHODS: Sprague-Dawley timed-mated rats were injected with flutamide (75 mg/kg body weight/5% ethanol + oil) on E16-19 or vehicle alone. Male fetuses or pups (5-10/group) were collected at E16; E19; and postnatal (P) days 0, 2, 4, 8. Sections were prepared for hematoxylin and eosin or immunohistochemistry for AR, ERα, and ERß. Receptor labeling was quantitated as distinct nuclear labeling/100 µm(2) in gubernaculum and IFP. RESULTS: There was minimal gubernacular AR-labeling until E19, dramatically increasing postnatally. By contrast, at E16-E19 there was significant IFP AR immunoreactivity suppressed by flutamide (P < .05). No ERα expression was observed, but ERß was expressed in both gubernaculum and IFP, maximally at E16, but unchanged by flutamide. CONCLUSIONS: During the androgen sensitivity window (E16-19), the gubernaculum contains ERß but minimal ERα or AR, while the IFP, which is supplied by the genitofemoral nerve, contains abundant AR that are flutamide-sensitive. These results suggest that the IFP could be the site of androgenic action controlling gubernacular development.


Subject(s)
Androgen Antagonists/pharmacology , Estrogen Receptor alpha/drug effects , Estrogen Receptor beta/drug effects , Flutamide/pharmacology , Intra-Abdominal Fat/drug effects , Ligaments/drug effects , Receptors, Androgen/drug effects , Testis/embryology , Animals , Cell Nucleus/chemistry , Cryptorchidism/physiopathology , Estrogen Receptor alpha/biosynthesis , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/biosynthesis , Estrogen Receptor beta/genetics , Female , Femoral Nerve/physiology , Gene Expression Regulation, Developmental/drug effects , Gestational Age , Intra-Abdominal Fat/embryology , Intra-Abdominal Fat/growth & development , Intra-Abdominal Fat/innervation , Intra-Abdominal Fat/metabolism , Ligaments/embryology , Ligaments/growth & development , Ligaments/metabolism , Male , Mammary Glands, Animal/embryology , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/metabolism , Pregnancy , Random Allocation , Rats , Rats, Sprague-Dawley , Receptors, Androgen/biosynthesis , Receptors, Androgen/genetics , Scrotum/embryology , Scrotum/growth & development , Testis/growth & development , Testosterone/physiology
12.
J Neurotrauma ; 27(1): 275-85, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19772460

ABSTRACT

Cardiometabolic risk factors are sorely underreported after spinal cord injury (SCI), despite the high prevalence of metabolic disorders and cardiovascular mortality in this population. Body-composition analysis and serum-lipid profiling are two assessments that are beginning to be more widely used to document metabolic changes after clinical SCI. Individuals with SCI have been reported to carry increased visceral fat and to exhibit altered serum-lipid levels. However, little is known about the development of these cardiometabolic risk factors in animal models. Using a combination of magnetic resonance imaging (MRI) and adipose tissue dissection, we show that visceral and subcutaneous adipose tissue were both increased at 1 month, but not at 1 week, after complete T3 SCI in rats. Additionally, at 1 month post injury, T3 SCI rats exhibited nonfasting serum hypertriglyceridemia, a result obtained using both standard clinical methods and a home cholesterol monitoring device (CardioChek). Interestingly, at 1 month post injury, rats with complete T10 SCI did not show an increase in either visceral adiposity or serum triglyceride levels. The fact that complete high-thoracic SCI disrupts lipid metabolism and perturbs fat storage in the subacute period, while low-thoracic SCI does not, suggests that differences in descending sympathetic control of adipose tissue might play a role in these changes. These results provide the first evidence of cardiometabolic risk factors in experimental animals with SCI, and are a starting point for investigations of the etiology of obesity and metabolic dysfunctions that often accompany SCI.


Subject(s)
Cardiovascular Diseases/etiology , Cardiovascular Diseases/metabolism , Lipid Metabolism Disorders/etiology , Lipid Metabolism Disorders/metabolism , Spinal Cord Injuries/complications , Spinal Cord Injuries/metabolism , Animals , Autonomic Nervous System Diseases/complications , Autonomic Nervous System Diseases/physiopathology , Cardiovascular Diseases/physiopathology , Disease Models, Animal , Dissection , Hyperlipidemias/etiology , Hyperlipidemias/metabolism , Hyperlipidemias/physiopathology , Intra-Abdominal Fat/innervation , Intra-Abdominal Fat/metabolism , Intra-Abdominal Fat/physiopathology , Lipid Metabolism/physiology , Lipid Metabolism Disorders/physiopathology , Magnetic Resonance Imaging , Male , Rats , Rats, Wistar , Risk Factors , Spinal Cord Injuries/physiopathology , Triglycerides/blood
13.
Spine (Phila Pa 1976) ; 30(13): 1501-4, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15990663

ABSTRACT

STUDY DESIGN: An experimental study in the pig with autologous transfer of nucleus pulpous and retroperitoneal fat to the subcutaneous space of the back. OBJECTIVES: To evaluate if there is neovascularization or neoinnervation of subcutaneously placed nucleus pulposus, in comparison to retroperitoneal fat, and under simultaneous treatment by certain antiangiogenetic drugs. SUMMARY OF BACKGROUND DATA: It has been suggested that intervertebral discs may be invaded by newly formed blood vessels and nerve fibers following injury of the anulus fibrosus. The nerve fibers have been considered to induce low back pain. However, it is still debated whether such ingrowth may occur and, if present, if this is based on the action of angiogenetic substances in the intervertebral disc or simply by normal would healing. METHODS: In the first series, autologous nucleus pulposus and retroperitoneal fat was placed subcutaneously in 3 pigs. In the second series, autologous nucleus pulposus was placed subcutaneously with simultaneous treatment with methotrexate (n = 3), celecoxib (n = 3), doxycycline (n = 3), and infliximab (n = 3). After 7 days, the tissue was collected and processed immunohistochemically for the visualization of blood vessels and nerve fibers. RESULTS: There was a number of blood vessels and nerve fibers in the nucleus pulposus samples, while no vessels were observed in the fat samples. Neither methotrexate nor celecoxib seemed to be able to reduce the ingrowth of blood vessels (neovascularization) or nerve fibers (neoinnervation). Treatment by doxycycline and infliximab markedly reduced both neovascularization and neoinnervation. CONCLUSIONS: Subcutaneously placed autologous nucleus pulposus displays an ingrowth of newly formed blood vessels and nerve fibers within 7 days, in contrast to retroperitoneal fat. Such ingrowth seems to be reduced by doxycycline and infliximab, 2 cytokine inhibitors. The data suggest that the ingrowth may be induced by bioactive substances within the nucleus pulposus. The clinical importance of these data has yet to be elucidated.


Subject(s)
Enzyme Inhibitors/pharmacology , Intervertebral Disc , Methotrexate/pharmacology , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/physiology , Nerve Regeneration/drug effects , Nerve Regeneration/physiology , Animals , Anti-Bacterial Agents/pharmacology , Antibodies, Monoclonal/pharmacology , Celecoxib , Cyclooxygenase Inhibitors/pharmacology , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Dermatologic Agents/pharmacology , Doxycycline/pharmacology , Infliximab , Intervertebral Disc/blood supply , Intervertebral Disc/innervation , Intervertebral Disc/transplantation , Intra-Abdominal Fat/blood supply , Intra-Abdominal Fat/innervation , Intra-Abdominal Fat/transplantation , Prostaglandins/metabolism , Pyrazoles/pharmacology , Subcutaneous Tissue , Sulfonamides/pharmacology , Swine , Transplantation, Autologous , Wound Healing/drug effects , Wound Healing/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...