Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 955
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 22(5): 639-653, 2021 05.
Article in English | MEDLINE | ID: mdl-33907320

ABSTRACT

White adipose tissue (WAT) is an essential regulator of energy storage and systemic metabolic homeostasis. Regulatory networks consisting of immune and structural cells are necessary to maintain WAT metabolism, which can become impaired during obesity in mammals. Using single-cell transcriptomics and flow cytometry, we unveil a large-scale comprehensive cellular census of the stromal vascular fraction of healthy lean and obese human WAT. We report new subsets and developmental trajectories of adipose-resident innate lymphoid cells, dendritic cells and monocyte-derived macrophage populations that accumulate in obese WAT. Analysis of cell-cell ligand-receptor interactions and obesity-enriched signaling pathways revealed a switch from immunoregulatory mechanisms in lean WAT to inflammatory networks in obese WAT. These results provide a detailed and unbiased cellular landscape of homeostatic and inflammatory circuits in healthy human WAT.


Subject(s)
Immunity, Innate , Obesity/immunology , Subcutaneous Fat, Abdominal/immunology , Abdominoplasty , Adipocytes/immunology , Adipocytes/metabolism , Adult , Cell Communication/immunology , Cell Line , Dendritic Cells, Follicular/immunology , Dendritic Cells, Follicular/metabolism , Female , Humans , Inflammation/immunology , Inflammation/pathology , Lymphocytes/immunology , Lymphocytes/metabolism , Macrophages/immunology , Macrophages/metabolism , Obesity/pathology , Obesity/surgery , RNA-Seq , Signal Transduction/immunology , Single-Cell Analysis , Subcutaneous Fat, Abdominal/pathology , Subcutaneous Fat, Abdominal/surgery
2.
Int J Obes (Lond) ; 48(4): 523-532, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38135702

ABSTRACT

BACKGROUND AND AIMS: Obesity predisposes to metabolic and cardiovascular diseases. Adipose tissue inflammation and systemic inflammation contribute to these complications. There are strong sex differences in adipose tissue distribution and in systemic inflammation. Women have more subcutaneous adipose tissue (SAT) and less visceral adipose tissue (VAT) than men. We explored the sex differences in the association between the different adipose compartments and inflammatory markers that are important in cardiometabolic disease pathophysiology. METHODS: Single-center observational cohort study with 302 individuals with a BMI ≥ 27 kg/m2. We were unable to acquire MRI data from seven individuals and from another 18 the MRI data were not usable, resulting in 277 people (155 men, 122 women), aged 55-81 years. INTERVENTION: We performed the following measurements: abdominal magnetic resonance imaging to measure VAT, and SAT (deep and superficial) volumes; circulating leukocyte counts and cytokine production capacity of peripheral blood mononuclear cells (PBMCs), circulating cytokines, adipokines, and targeted proteomics; abdominal sSAT biopsies for histology and gene expression. RESULTS: Only in women, (s)SAT volume was associated with circulating leukocytes, monocytes, and neutrophils. Circulating IL-6 and IL-18BP were associated with SAT volume in women and VAT in men. Several circulating proteins, including monocyte-colony-stimulating factor 1 and hepatocyte growth factor, are associated with sSAT in women and VAT in men. Only in women, SAT volume is associated with SAT expression of inflammatory proteins, including leptin, CD68, TNFα and IL-1α. CONCLUSION: In women living with obesity, abdominal SAT volume, especially sSAT, is associated with circulating leukocytes and inflammatory proteins. In men, these parameters mainly show associations with VAT volume. This could be because only in women, sSAT volume is associated with sSAT expression of inflammatory proteins. These findings underscore that future research on adipose tissue in relation to cardiometabolic and cardiovascular disease should take sex differences into account.


Subject(s)
Cardiovascular Diseases , Leukocytes, Mononuclear , Humans , Female , Male , Leukocytes, Mononuclear/metabolism , Obesity/metabolism , Subcutaneous Fat/metabolism , Inflammation/metabolism , Adipose Tissue/metabolism , Subcutaneous Fat, Abdominal/metabolism , Cardiovascular Diseases/complications , Immunity, Innate , Intra-Abdominal Fat/metabolism
3.
BMC Public Health ; 24(1): 1827, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38982435

ABSTRACT

BACKGROUND: Cardiovascular diseases (CVDs) are the primary cause of mortality globally. The prevalence of obesity is rising worldwide; there seems to be a significant positive association between obesity and CVDs. The distribution of fat in the abdominal area in the form of visceral (VAT) or subcutaneous adipose tissue (SAT) affects the risk of CVDs. The aim of the present study was to conduct a systematic review of the available literature regarding the association between the VAT-to-SAT ratio and CVDs. METHODS: A comprehensive search strategy was used to retrieve all human observational studies indexed in PubMed, Scopus and Google Scholar databases/search engines (from Jan 2000 up to Oct 2023). The VAT-to-SAT or SAT-to-VAT ratio was an independent variable and various cardiovascular diseases, including hypertension, atherosclerosis, coronary heart disease, cerebrovascular disease and heart failure, were considered as outcomes of interest. RESULTS: Out of 1173 initial studies, 910 papers were screened. Based on the inclusion criteria, 883 papers were excluded. Finally, 27 papers (18 cross-sectional and 9 cohort studies) published between 2010 and 2023 which met the inclusion criteria were reviewed. CONCLUSIONS: The distribution of abdominal fat seems to be associated with the risk of CVDs; the majority of the evidence suggests that a higher abdominal VAT-to-SAT ratio is associated with the development of CVDs. Therefore, this ratio can be used as a prognostic indicator for CVDs. TRIAL REGISTRATION: Not applicable.


Subject(s)
Cardiovascular Diseases , Intra-Abdominal Fat , Subcutaneous Fat, Abdominal , Humans , Cardiovascular Diseases/epidemiology , Risk Factors
4.
J Obstet Gynaecol ; 44(1): 2329880, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38516715

ABSTRACT

BACKGROUND: This investigation aimed to analyse the efficacy of abdominal subcutaneous fat thickness (ASFT) value >18.1 mm combined with a 50-g glucose challenge test (GCT) between 24-28 weeks of gestation in predicting gestational diabetes mellitus (GDM) cases. METHODS: This cross-sectional study was carried out from February 2021 to December 2022. All pregnant women received a 50-g GCT at 24-28 weeks of pregnancy for the GDM screening. Pregnant women with a blood glucose value between 140-190 mg/dl experienced 100 g OGTT. Even if 50-g GCT was normal, 100-g OGTT was offered to patients with an ASFT value above 18.1 mm. RESULTS: Among the 728 pregnant women we enrolled, 154 (21.2%) cases were screened as positive. The number of patients who first screened positive and determined to be GDM after the 100-g oral glucose tolerance test (OGTT) was 43 (5.9%). A total of 67 cases (9.2%) had an ASFT measurement above 18.1 mm. Two cases with a negative 50-g GCT and ASFT <18.1 mm were diagnosed as GDM in the later weeks of pregnancy. A 50-g GCT combined with ASFT measurement above 18.1 mm predicted GDM with a sensitivity of 87.9%, a specificity of 88.7%, a positive predictive value (PPV) of 36.0%, and a negative PV (NPV) of 99.7%. CONCLUSIONS: A 50-g GCT combined with ASFT measurement that can be easily and accurately obtained during routine antenatal care in the second trimester might be a beneficial indicator for predicting GDM cases.


Screening and diagnosing pregnant women at greater risk of developing gestational diabetes mellitus are crucial to enhancing short- and long-term outcomes of the mother and foetus. An accurate diagnosis could provide proper treatment, which could be dietary or pharmacological, manage the disease, and improve pregnancy outcomes. In the current study, we revealed that gestational diabetes was predicted with high sensitivity and specificity in pregnant women with a 50-gram glucose challenge test and abdominal subcutaneous fat thickness measurement above 18.1 millimetres. Therefore, abdominal subcutaneous fat thickness measurement is anticipated to be extensively used as an indicative variable for predicting gestational diabetes mellitus cases during the second trimester of pregnancy.


Subject(s)
Diabetes, Gestational , Pregnancy , Female , Humans , Diabetes, Gestational/diagnosis , Glucose Tolerance Test , Subcutaneous Fat, Abdominal , Cross-Sectional Studies , Blood Glucose
5.
Acta Radiol ; 64(5): 1770-1774, 2023 May.
Article in English | MEDLINE | ID: mdl-36748104

ABSTRACT

BACKGROUND: Patients with underlying hematologic malignancy have a higher risk of developing systemic amyloidosis, which worsens their prognosis. Histopathologic detection of amyloid deposits in tissue biopsy specimens is the only diagnostic method for amyloidosis. PURPOSE: To compare the efficacy of ultrasound-guided percutaneous core needle biopsy (USPCB) of abdominal subcutaneous fat with that of bone marrow biopsy (BMB) for diagnosing amyloidosis. MATERIAL AND METHODS: A total of 90 consecutive patients with underlying hematologic disorders who underwent both USPCB of abdominal subcutaneous fat and BMB for suspicion of amyloid deposition during a 10-year period were included in this retrospective study. RESULTS: The sensitivity and specificity of detecting amyloid deposition were 85.7% and 100%, respectively, with USPCB as opposed to 4.8% and 100%, respectively, with BMB, and the sensitivity was significantly higher with USPCB (P < 0.001). The mean number of times USPCB was performed was 3.3. There were no major complications associated with USPCB. The sensitivity of detecting amyloidosis was not different between the 18-G needle group and the 14-G group (100% vs. 80%; P = 0.623). Logistic regression analysis revealed that acquiring more cores from USPCB and thinner fat tissues were statistically significant factors that affected the diagnostic accuracy of USPCB for amyloid detection. CONCLUSION: The sensitivity of amyloid deposition was significantly higher with USPCB of abdominal subcutaneous fat than BMB. Acquiring more cores by multiple biopsies instead of using a larger bore needle and thin subcutaneous fat pad may be a favorable factor for the diagnostic accuracy of USPCB.


Subject(s)
Amyloidosis , Subcutaneous Fat, Abdominal , Humans , Biopsy, Large-Core Needle , Subcutaneous Fat, Abdominal/pathology , Retrospective Studies , Bone Marrow/diagnostic imaging , Bone Marrow/pathology , Biopsy , Amyloidosis/diagnostic imaging , Image-Guided Biopsy , Ultrasonography, Interventional
6.
Gastroenterology ; 161(3): 968-981.e12, 2021 09.
Article in English | MEDLINE | ID: mdl-34004161

ABSTRACT

BACKGROUND AND AIMS: Insulin resistance is a key factor in the pathogenesis of nonalcoholic fatty liver disease (NAFLD). We evaluated the importance of subcutaneous abdominal adipose tissue (SAAT) inflammation and both plasma and SAAT-derived exosomes in regulating insulin sensitivity in people with obesity and NAFLD. METHODS: Adipose tissue inflammation (macrophage and T-cell content and expression of proinflammatory cytokines), liver and whole-body insulin sensitivity (assessed using a hyperinsulinemic-euglycemic clamp and glucose tracer infusion), and 24-hour serial plasma cytokine concentrations were evaluated in 3 groups stratified by adiposity and intrahepatic triglyceride (IHTG) content: (1) lean with normal IHTG content (LEAN; N = 14); (2) obese with normal IHTG content (OB-NL; N = 28); and (3) obese with NAFLD (OB-NAFLD; N = 28). The effect of plasma and SAAT-derived exosomes on insulin-stimulated Akt phosphorylation in human skeletal muscle myotubes and mouse primary hepatocytes was assessed in a subset of participants. RESULTS: Proinflammatory macrophages, proinflammatory CD4 and CD8 T-cell populations, and gene expression of several cytokines in SAAT were greater in the OB-NAFLD than the OB-NL and LEAN groups. However, with the exception of PAI-1, which was greater in the OB-NAFLD than the LEAN and OB-NL groups, 24-hour plasma cytokine concentration areas-under-the-curve were not different between groups. The percentage of proinflammatory macrophages and plasma PAI-1 concentration areas-under-the-curve were inversely correlated with both hepatic and whole-body insulin sensitivity. Compared with exosomes from OB-NL participants, plasma and SAAT-derived exosomes from the OB-NAFLD group decreased insulin signaling in myotubes and hepatocytes. CONCLUSIONS: Systemic insulin resistance in people with obesity and NAFLD is associated with increased plasma PAI-1 concentrations and both plasma and SAAT-derived exosomes. ClinicalTrials.gov number: NCT02706262 (https://clinicaltrials.gov/ct2/show/NCT02706262).


Subject(s)
Cytokines/blood , Exosomes/metabolism , Insulin Resistance , Macrophages/metabolism , Memory T Cells/metabolism , Non-alcoholic Fatty Liver Disease/blood , Obesity/blood , Plasminogen Activator Inhibitor 1/blood , Subcutaneous Fat, Abdominal/metabolism , Adult , Animals , Biomarkers/blood , Blood Glucose/metabolism , Cells, Cultured , Exosomes/immunology , Female , Hepatocytes/metabolism , Humans , Insulin/blood , Liver/metabolism , Macrophages/immunology , Male , Memory T Cells/immunology , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/metabolism , Non-alcoholic Fatty Liver Disease/diagnosis , Non-alcoholic Fatty Liver Disease/immunology , Non-alcoholic Fatty Liver Disease/physiopathology , Obesity/diagnosis , Obesity/immunology , Obesity/physiopathology , Subcutaneous Fat, Abdominal/immunology , Tissue Culture Techniques
7.
Hepatology ; 74(3): 1287-1299, 2021 09.
Article in English | MEDLINE | ID: mdl-33743554

ABSTRACT

BACKGROUND AND AIMS: It is proposed that impaired expansion of subcutaneous adipose tissue (SAT) and an increase in adipose tissue (AT) fibrosis causes ectopic lipid accumulation, insulin resistance (IR), and metabolically unhealthy obesity. We therefore evaluated whether a decrease in SAT expandability, assessed by measuring SAT lipogenesis (triglyceride [TG] production), and an increase in SAT fibrogenesis (collagen production) are associated with NAFLD and IR in persons with obesity. APPROACH AND RESULTS: In vivo abdominal SAT lipogenesis and fibrogenesis, expression of SAT genes involved in extracellular matrix (ECM) formation, and insulin sensitivity were assessed in three groups of participants stratified by adiposity and intrahepatic TG (IHTG) content: (1) healthy lean with normal IHTG content (Lean-NL; n = 12); (2) obese with normal IHTG content and normal glucose tolerance (Ob-NL; n = 25); and (3) obese with NAFLD and abnormal glucose metabolism (Ob-NAFLD; n = 25). Abdominal SAT TG synthesis rates were greater (P < 0.05) in both the Ob-NL (65.9 ± 4.6 g/wk) and Ob-NAFLD groups (71.1 ± 6.7 g/wk) than the Lean-NL group (16.2 ± 2.8 g/wk) without a difference between the Ob-NL and Ob-NAFLD groups. Abdominal SAT collagen synthesis rate and the composite expression of genes encoding collagens progressively increased from the Lean-NL to the Ob-NL to the Ob-NAFLD groups and were greater in the Ob-NAFLD than the Ob-NL group (P < 0.05). Composite expression of collagen genes was inversely correlated with both hepatic and whole-body insulin sensitivity (P < 0.001). CONCLUSIONS: AT expandability is not impaired in persons with obesity and NAFLD. However, SAT fibrogenesis is greater in persons with obesity and NAFLD than in those with obesity and normal IHTG content, and is inversely correlated with both hepatic and whole-body insulin sensitivity.


Subject(s)
Collagen/metabolism , Glucose Intolerance/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/metabolism , Subcutaneous Fat, Abdominal/metabolism , Triglycerides/metabolism , Adipose Tissue/metabolism , Adult , Extracellular Matrix/metabolism , Female , Fibrosis , Glucose Intolerance/complications , Humans , Insulin Resistance , Lipogenesis , Male , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Non-alcoholic Fatty Liver Disease/complications , Obesity/complications , Subcutaneous Fat/metabolism
8.
Dermatol Ther ; 35(9): e15717, 2022 09.
Article in English | MEDLINE | ID: mdl-35837791

ABSTRACT

Various treatment methods are used for noninvasive body contouring. To evaluate the efficacy and safety of a newly designed cryolipolysis device using a three-dimensional cooling method for abdominal fat reduction. Twenty-five participants with clinically apparent abdominal fat tissue participated in the study. The thickness of fat tissue below the umbilicus level was measured using a caliper at baseline and 12 weeks after the first treatment. The height of abdominal subcutaneous fat tissue on ultrasonography and participant satisfaction were assessed at every visit for 16 weeks. All adverse events (AEs) during the study period were recorded. p values <0.05 were considered statistically significant. Twenty-four participants completed this study; the mean BMI of participants was 29.34 ± 2.36 kg/m2 . The mean thickness of abdominal subcutaneous fat was significantly lower at 12 weeks (40.4 ± 6.8 mm, p < 0.001) than at baseline (49.3 ± 8.5 mm). Differences in the height of abdominal subcutaneous fat compared to that at baseline were 1.02 ± 0.41 cm (12 weeks, p < 0.001) and 1.13 ± 0.44 cm (16 weeks, p < 0.001). Rates of abdominal subcutaneous fat reduction at 12 and 16 weeks compared to that at baseline were 28.45% and 31.13%, respectively. The ratio of abdominal circumference to hip circumference at 12 and 16 weeks was significantly decreased compared to that at baseline. Most participants (95.8%) reported improvement in satisfaction scores at 16 weeks. There were no serious AEs during the entire study period. The study demonstrated the efficacy of a noninvasive cryolipolysis device using a three-dimensional cooling method for reducing abdominal subcutaneous fat.


Subject(s)
Body Contouring , Lipectomy , Body Contouring/adverse effects , Body Contouring/methods , Humans , Lipectomy/adverse effects , Lipectomy/methods , Patient Satisfaction , Prospective Studies , Subcutaneous Fat, Abdominal/diagnostic imaging , Subcutaneous Fat, Abdominal/surgery , Treatment Outcome
9.
Lasers Med Sci ; 37(1): 505-512, 2022 Feb.
Article in English | MEDLINE | ID: mdl-33797649

ABSTRACT

The aim of this study was to evaluate the safety and efficacy of combined 1060-nm diode laser and 635-nm low-level laser therapy (LLLT) device for non-invasive reduction of the abdominal and submental fat. Forty-two healthy subjects received single laser treatment on both the abdomen and submental area. Ultrasound images measuring the thickness of abdominal and submental fat were taken at baseline, follow-up at 4, 8, and 12 weeks after treatment. Waist circumference and body weight were also measured at all visits. Adverse events were recorded at all visits. Subjects completed a satisfaction questionnaire at the end of the trial. Twelve weeks after a single treatment with the investigational device, ultrasound images showed statistically significant (P < 0.0001) reductions in abdominal and submental fat by 18.62 and 26.4%, respectively. In addition, significant (P < 0.0001) reduction in waist circumference was observed. Ninety-six percent of subjects rated that they were satisfied. Noted side effects were transient mild to moderate tenderness which subsided within 1 to 3 weeks. No serious treatment-related adverse events were reported. The dual wavelength device combining 1060-nm diode laser with 635-nm LLLT was safe and effective for non-invasive reduction of both abdominal and submental fat.


Subject(s)
Lasers, Semiconductor , Lipectomy , Low-Level Light Therapy , Combined Modality Therapy/adverse effects , Humans , Lasers, Semiconductor/adverse effects , Lipectomy/instrumentation , Low-Level Light Therapy/instrumentation , Subcutaneous Fat/diagnostic imaging , Subcutaneous Fat/surgery , Subcutaneous Fat, Abdominal/diagnostic imaging , Subcutaneous Fat, Abdominal/surgery , Treatment Outcome , Ultrasonography
10.
Int J Mol Sci ; 23(4)2022 Feb 21.
Article in English | MEDLINE | ID: mdl-35216509

ABSTRACT

The metabolic syndrome (MetS) is a cluster of the most dangerous heart attack risk factors: diabetes or raised fasting plasma glucose, abdominal obesity, high cholesterol and high blood pressure. The goal of this study is to compare the state of the main features of obesity-associated white adipose tissue (WAT) dysfunction in 66 women with severe obesity without (MetS-) or with MetS (MetS+). Fat cell area, adipocyte size distribution and histological fibrosis were analysed in visceral (VAT) and abdominal subcutaneous WAT (SAT) in 33 age- and BMI-matched pairs of MetS- and MetS+ subjects. The mRNA expression of 93 genes implicated in obesity-associated WAT dysfunction was analysed by RT-qPCR in both fat depots. MetS+ females showed higher adipocyte hypertrophy in both fat depots and increased fibrosis and expression of macrophage and hypoxia markers in SAT. Transcriptional data suggest increased fatty acid oxidation in SAT and impaired thermogenesis and extracellular matrix remodelling in VAT from MetS+ subjects. A sPLS-DA model, including SAT expression of PPARA and LEPR genes identified MetS with an AUC = 0.87. Despite equal age, BMI and body composition, MetS+ females display morphological and transcriptional differences in both WAT depots, especially in SAT. These factors may contribute to the transition to MetS.


Subject(s)
Metabolic Syndrome/pathology , Obesity, Morbid/pathology , Subcutaneous Fat, Abdominal/pathology , Adipocytes/metabolism , Adipocytes/pathology , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Body Composition/physiology , Body Mass Index , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Female , Humans , Hypertension/metabolism , Hypertension/pathology , Intra-Abdominal Fat/metabolism , Metabolic Syndrome/metabolism , Middle Aged , Obesity, Abdominal/metabolism , Obesity, Abdominal/pathology , Obesity, Morbid/metabolism , Subcutaneous Fat, Abdominal/metabolism , Thermogenesis/physiology
11.
Aesthet Surg J ; 42(11): NP662-NP674, 2022 10 13.
Article in English | MEDLINE | ID: mdl-35709509

ABSTRACT

BACKGROUND: CBL-514 is a novel injectable drug that may be safe and efficacious for localized abdominal subcutaneous fat reduction. OBJECTIVES: The aim of this study was to assess the safety and efficacy of CBL-514 in reducing abdominal subcutaneous fat volume and thickness. METHODS: This Phase IIa, open-label, random allocation study consisted of a 6-week treatment period and follow-up at 4 and 8 weeks following the last treatment. Participants were randomly allocated to receive 1.2 mg/cm2 (180 mg), 1.6 mg/cm2 (240 mg), or 2.0 mg/cm2 (300 mg) of CBL-514 with up to 4 treatments, each comprising 60 injections into the abdominal adipose layer. Changes in abdominal subcutaneous fat were assessed by ultrasound at follow-up visits. Treatment-emergent adverse events were recorded. RESULTS: Higher doses of CBL-514 (unit dose, 2.0 and 1.6 mg/cm2) significantly improved the absolute and percentage reduction in abdominal fat volume (P < 0.00001) and thickness (P < 0.0001) compared with baseline. Although the COVID-19 pandemic halted some participant recruitment and follow-ups, analysis was unaffected, even after sample size limitations. CONCLUSIONS: CBL-514 injection at multiple doses up to 300 mg with a unit dose of 2.0 mg/cm2 is safe, well-tolerated, and reduced abdominal fat volume and thickness by inducing adipocyte apoptosis. Although other procedures exist to treat abdominal fat, they have limitations and may cause complications. At a dose of 2.0 mg/cm2, CBL-514 safely and significantly reduced abdominal fat volume by 24.96%, making it a promising new treatment for routine, nonsurgical abdominal fat reduction in dermatologic clinics.


Subject(s)
COVID-19 , Subcutaneous Fat, Abdominal , Adipocytes , Apoptosis , Humans , Lipolysis , Pandemics , Subcutaneous Fat/diagnostic imaging , Subcutaneous Fat, Abdominal/diagnostic imaging , Subcutaneous Fat, Abdominal/surgery , Treatment Outcome
12.
BMC Cancer ; 21(1): 1164, 2021 Oct 30.
Article in English | MEDLINE | ID: mdl-34715813

ABSTRACT

BACKGROUND: The incidence of hepatocellular carcinoma (HCC) has been increasing among the elderly populations. Trans-arterial chemoembolization (TACE), a widely used first-line non-curative therapy for HCCs is an issue in geriatrics. We investigated the prognosis of elderly HCC patients treated with TACE and determined the factors that affect the overall survival. METHODS: We included 266 patients who were older than 65 years and had received TACE as initial treatment for HCC. We analyzed the skeletal muscle index (SMI) and visceral-to-subcutaneous fat ratio (VSR) around the third lumbar vertebrae using computed tomography scans. Muscle depletion with visceral adiposity (MDVA) was defined by falling below the median SMI and above the median VSR value sex-specifically. We evaluated the overall survival in association with MDVA and other clinical factors. RESULTS: The mean age was 69.9 ± 4.5 years, and 70.3% of the patients were men. According to the Barcelona Clinic Liver Cancer (BCLC) staging system, 29, 136, and 101 patients were classified as BCLC 0, A, and B stages, respectively, and 79 (29.7%) had MDVA. During the median follow-up of 4.1 years, patients with MDVA had a shorter life expectancy than those without MDVA (P = 0.007) even though MDVA group had a higher objective response rate after the first TACE (82.3% vs. 75.9%, P = 0.035). Multivariate analysis revealed that MDVA (Hazard ratio [HR] 1.515) age (HR 1.057), liver function (HR 1.078), tumor size (HR 1.083), serum albumin level (HR 0.523), platelet count (HR 0.996), tumor stage (stage A, HR 1.711; stage B, HR 2.003), and treatment response after the first TACE treatment (HR 0.680) were associated with overall survival. CONCLUSIONS: MDVA is a critical prognostic factor for predicting survival in the elderly patients with HCC who have undergone TACE.


Subject(s)
Carcinoma, Hepatocellular/mortality , Chemoembolization, Therapeutic , Intra-Abdominal Fat , Liver Neoplasms/mortality , Sarcopenia/mortality , Subcutaneous Fat, Abdominal , Adiposity , Aged , Body Composition , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Female , Humans , Intra-Abdominal Fat/diagnostic imaging , Life Expectancy , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Male , Muscle, Skeletal/diagnostic imaging , Neoplasm Staging , Prognosis , Republic of Korea , Retrospective Studies , Sarcopenia/diagnostic imaging , Subcutaneous Fat, Abdominal/diagnostic imaging
13.
J Theor Biol ; 530: 110883, 2021 12 07.
Article in English | MEDLINE | ID: mdl-34478744

ABSTRACT

BACKGROUND: A good treatment for type 1 diabetes mellitus (T1DM) requires accurate measurements of blood glucose levels. Continuous glucose monitors (CGM) measure the glucose concentration in the interstitial fluid of the abdominal subcutaneous adipose tissue. However, glucose measured in the abdominal interstitial fluid does not represent blood glucose concentrations accurately due to the complex blood transport through the body and glucose diffusion in interstitial fluid. METHODS: To gain insight into this problem, a phenomenological-based semiphysical model (PBSM) of glucose transport by volumetric flow and diffusion from the bloodstream to interstitial fluid was constructed. A published 10-step modeling procedure was used to obtain a model for glucose transport time through the blood vessels and from the blood capillaries to the interstitial fluid, glucose diffusion within the interstitial fluid, and glucose diffusion through the semipermeable coating of the sensor needle. For this model, a healthy person is considered at rest with average parameters. RESULTS: The simulations performed using the PBSM allow obtaining the glucose transport time from the liver to the sensor needle. In this way, it is possible to reconstruct an accurate dynamic measurement of blood glucose from the measurements in the interstitial fluid of the abdominal subcutaneous adipose tissue. CONCLUSIONS: PBSMs with parameters interpretability illustrate the connection of glucose concentrations in the interstitial fluid with that currently in the blood. Implementing this model in a CGM will result in more reliable measurements of blood glucose levels for T1DM treatment.


Subject(s)
Diabetes Mellitus, Type 1 , Subcutaneous Fat, Abdominal , Adipose Tissue , Blood Glucose , Glucose , Humans , Liver
14.
BMC Pregnancy Childbirth ; 21(1): 490, 2021 Jul 07.
Article in English | MEDLINE | ID: mdl-34233642

ABSTRACT

BACKGROUND: It was reported that steroid-related gene expressions in the adipose tissue (AT) of women differ between women affected with polycystic ovary syndrome (PCOS) and non-PCOS. Although association between PCOS in mother and offspring's health is a crucial issue, there are few studies focusing on AT of pregnant women suffering from PCOS. Our objectives were to determine the differences between mRNA expression levels of key steroid-converting enzymes in abdominal subcutaneous AT of pregnant women afflicted with PCOS and non-PCOS. METHODS: Twelve pregnant women with PCOS (case) and thirty six non-PCOS pregnant women (control) (1:3 ratio; age- and BMI-matched) undergoing cesarean section were enrolled for the present study. Expressions of fifteen genes related to steriodogenesis in abdominal subcutaneous AT were investigated using quantitative real-time PCR. RESULTS: No significant differences were detected with respect to age, BMI (prior pregnancy and at delivery day), gestational period and parity among pregnant women with PCOS and non-PCOS. Most of the sex steroid-converting genes except 17ß-Hydroxysteroid dehydrogenases2 (17BHSD2), were highly expressed on the day of delivery in subcutaneous AT. Women with PCOS showed significantly higher mRNA levels of steroidgenic acute regulator (STAR; P < 0.001), cytochrome P450 monooxygenase (CYP11A1; P < 0.05), 17α-hydroxylase (CYP17A1; P < 0.05), and 11ß-Hydroxysteroid dehydrogenase (11BHSD1 and 11BHSD2; P < 0.05). The expression of steroid 21-hydroxylase (CYP21) in non-PCOS was fourfold higher than those of women with PCOS (P < 0.001). There were no significant differences between relative expression of aromatase cytochrome P450 (CYP19A1), 3ß-hydroxysteroid dehydrogenase (3BHSD1 and 3BHSD2), and 17BHSD family (1, 3, 5, 7, and 12) between the two groups. CONCLUSION: The expression levels of genes related to sex steroids metabolism were similar to age-matched and BMI- matched pregnant non-PCOS and pregnant women with PCOS at delivery day. However, the alterations in gene expressions involved in glucocorticoids and mineralocorticoids metabolism were shown. It is necessary to point out that further studies regarding functional activity are required. More attention should be given to AT of pregnant women with PCOS that was previously ignored.


Subject(s)
Gonadal Steroid Hormones/metabolism , Hydroxysteroid Dehydrogenases/metabolism , Polycystic Ovary Syndrome/genetics , Steroid Hydroxylases/metabolism , Subcutaneous Fat, Abdominal/metabolism , Adult , Case-Control Studies , Cesarean Section , Female , Gene Expression/genetics , Glucocorticoids/metabolism , Humans , Mineralocorticoids/metabolism , Phosphoproteins/metabolism , Pregnancy , RNA, Messenger/metabolism
15.
Microsurgery ; 41(4): 341-347, 2021 May.
Article in English | MEDLINE | ID: mdl-33720454

ABSTRACT

INTRODUCTION: Patient selection for autologous tissue transfer for postmastectomy breast reconstruction often utilizes body mass index (BMI) to risk stratify patients, though it only estimates fat content and does not address fat distribution. This study aims to identify a measurement of abdominal subcutaneous fat thickness (ASFT) from preoperative computed tomography (CT) angiography imaging to better predict complications. METHODS: A retrospective review of patients who underwent an abdominal microvascular free flap breast reconstruction was performed. The average of the bilateral distances from the lateral border of the rectus abdominus to the most proximal point of the dermis at the L4-L5 space was measured on preoperative imaging to estimate ASFT. This measurement was compared to BMI in regards to correlation with any complication, major or minor complications, and donor or recipient site complications. Statistical analysis utilized point-biserial correlations and multivariable logistic regression analyses. RESULTS: Three hundred and nine cases comprising a total of 496 breast reconstructions were identified. BMI did not correlate with any of the grouped complications, while ASFT correlated with occurrence of any complication (p = .003), minor complications (p = .001), and recipient site complications (p = .001). Further analysis revealed ASFT is specifically correlated with fat necrosis (p = .005). In independent multivariable regression models, both BMI (p = .011) and ASFT (p = .001) were significant predictors of fat necrosis. The ASFT model had a BIC of 335.42 compared to the BMI model with a value of 340.89, with smaller numbers representing more predictive models. CONCLUSION: Estimation of ASFT is easily performed and is a significantly better predictor of flap fat necrosis than BMI.


Subject(s)
Breast Neoplasms , Mammaplasty , Body Mass Index , Breast Neoplasms/surgery , Female , Humans , Mammaplasty/adverse effects , Mastectomy , Postoperative Complications/diagnostic imaging , Postoperative Complications/epidemiology , Postoperative Complications/etiology , Retrospective Studies , Subcutaneous Fat, Abdominal/diagnostic imaging , Subcutaneous Fat, Abdominal/surgery
16.
Gastroenterology ; 156(6): 1742-1752, 2019 05.
Article in English | MEDLINE | ID: mdl-30677401

ABSTRACT

BACKGROUND & AIMS: Identifying metabolic abnormalities that occur before pancreatic ductal adenocarcinoma (PDAC) diagnosis could increase chances for early detection. We collected data on changes in metabolic parameters (glucose, serum lipids, triglycerides; total, low-density, and high-density cholesterol; and total body weight) and soft tissues (abdominal subcutaneous fat [SAT], adipose tissue, visceral adipose tissue [VAT], and muscle) from patients 5 years before the received a diagnosis of PDAC. METHODS: We collected data from 219 patients with a diagnosis of PDAC (patients) and 657 healthy individuals (controls) from the Rochester Epidemiology Project, from 2000 through 2015. We compared metabolic profiles of patients with those of age- and sex-matched controls, constructing temporal profiles of fasting blood glucose, serum lipids including triglycerides, cholesterol profiles, and body weight and temperature for 60 months before the diagnosis of PDAC (index date). To construct the temporal profile of soft tissue changes, we collected computed tomography scans from 68 patients, comparing baseline (>18 months before diagnosis) areas of SAT, VAT, and muscle at L2/L3 vertebra with those of later scans until time of diagnosis. SAT and VAT, isolated from healthy individuals, were exposed to exosomes isolated from PDAC cell lines and analyzed by RNA sequencing. SAT was collected from KRAS+/LSLG12D P53flox/flox mice with PDACs, C57/BL6 (control) mice, and 5 patients and analyzed by histology and immunohistochemistry. RESULTS: There were no significant differences in metabolic or soft tissue features of patients vs controls until 30 months before PDAC diagnosis. In the 30 to 18 months before PDAC diagnosis (phase 1, hyperglycemia), a significant proportion of patients developed hyperglycemia, compared with controls, without soft tissue changes. In the 18 to 6 months before PDAC diagnosis (phase 2, pre-cachexia), patients had significant increases in hyperglycemia and decreases in serum lipids, body weight, and SAT, with preserved VAT and muscle. In the 6 to 0 months before PDAC diagnosis (phase 3, cachexia), a significant proportion of patients had hyperglycemia compared with controls, and patients had significant reductions in all serum lipids, SAT, VAT, and muscle. We believe the patients had browning of SAT, based on increases in body temperature, starting 18 months before PDAC diagnosis. We observed expression of uncoupling protein 1 (UCP1) in SAT exposed to PDAC exosomes, SAT from mice with PDACs, and SAT from all 5 patients but only 1 of 4 controls. CONCLUSIONS: We identified 3 phases of metabolic and soft tissue changes that precede a diagnosis of PDAC. Loss of SAT starts 18 months before PDAC identification, and is likely due to browning. Overexpression of UCP1 in SAT might be a biomarker of early-stage PDAC, but further studies are needed.


Subject(s)
Cachexia/etiology , Carcinoma, Pancreatic Ductal/blood , Carcinoma, Pancreatic Ductal/diagnosis , Hyperglycemia/blood , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/diagnosis , Adipocytes/metabolism , Adipocytes/pathology , Animals , Blood Glucose/metabolism , Body Temperature , Body Weight , Carcinoma, Pancreatic Ductal/complications , Carcinoma, Pancreatic Ductal/genetics , Case-Control Studies , Cells, Cultured , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Exosomes , Humans , Hyperglycemia/etiology , Intra-Abdominal Fat/diagnostic imaging , Intra-Abdominal Fat/pathology , Mice , Middle Aged , Muscle, Skeletal/diagnostic imaging , Pancreatic Neoplasms/complications , Pancreatic Neoplasms/genetics , RNA, Messenger/metabolism , Retrospective Studies , Subcutaneous Fat, Abdominal/diagnostic imaging , Subcutaneous Fat, Abdominal/pathology , Time Factors , Tomography, X-Ray Computed , Triglycerides/blood , Uncoupling Protein 1/genetics , Up-Regulation
17.
J Intern Med ; 288(2): 219-233, 2020 08.
Article in English | MEDLINE | ID: mdl-32406570

ABSTRACT

OBJECTIVE: Patients undergoing bariatric surgery present long-term metabolic improvements and reduced type 2 diabetes risk, despite long-term weight regain. We hypothesized that part of these protective effects could be linked to altered gene expression in white adipose tissue (WAT). METHODS: Transcriptomic profiling by gene microarray was performed in abdominal subcutaneous WAT from women before (n = 50) and two (n = 49) and five (n = 38) years after Roux-en-Y gastric bypass (RYGB) surgery as well as in 28 age-matched nonoperated women. RESULTS: In the obese women, the average body weight decrease was 38 kg 2 years postsurgery followed by an 8 kg weight regain between 2 and 5 years. Most of the long-term changes in WAT gene expression occurred during the first 2 years. However, a subset of genes encoding proteins involved in inflammation displayed a continued decrease between baseline, 2 and 5 years, respectively; that is an expression pattern independent of body weight regain. Expression of 71 of these genes correlated with measurements of adipocyte morphology or serum adipokine levels. CONCLUSION: The continuous improvement in WAT inflammatory gene expression, despite body weight relapse, may contribute to the sustained effects on adipose morphology after bariatric surgery.


Subject(s)
Gastric Bypass , Gene Expression , Subcutaneous Fat, Abdominal/metabolism , Adipocytes , Adiponectin/blood , Adult , Body Mass Index , Case-Control Studies , Cell Count , Cell Size , Down-Regulation , Female , Follow-Up Studies , Gene Ontology , Humans , Leptin/blood , Middle Aged , Tissue Array Analysis , Up-Regulation
18.
Int J Obes (Lond) ; 44(2): 377-387, 2020 02.
Article in English | MEDLINE | ID: mdl-31164724

ABSTRACT

BACKGROUND: Transcriptome analysis of abdominal subcutaneous white adipose tissue (sWAT) has identified important obesity-associated disturbances. However, the relation between sWAT transcriptome and long-term future changes in body weight remains elusive. OBJECTIVE: To investigate sWAT transcriptome signatures before and after long-term weight changes and assess their predictive value for body weight changes. DESIGN: A total of 56 women were followed longitudinally and subdivided into weight-stable (WS, n = 25), weight-gaining (WG, n = 14) and weight-losing (WL, n = 17) groups between baseline and follow-up (13 ± 1 years). The fasting sWAT transcriptome was analyzed by gene microarray at baseline and follow-up. Key genes associated with weight changes were validated using quantitative real-time PCR. RESULTS: In total 285 transcripts exhibited difference (FDR < 30%) in expression fold change over time between WL and WS women. WL women displayed decreased pro-inflammatory (NLRP3) but increased insulin-response gene (FASN and GLUT4) expression over time. In comparison, 461 transcripts displayed difference in expression fold change over time between WG and WS women (P < 0.05). Genes involved in autophagic processes (CDK5, SQSTM1 and FBXL2) were generally upregulated in WG women. At baseline, 307 and 302 transcripts were differentially expressed (FDR < 30%) in WL and WG women, respectively, when independently compared against WS women. Baseline expression of adipogenic and lipogenic genes (PPARG, IRS2 and HACD2) was lower, while pro-fibrotic (COL6A1) was higher, in WL than WS women; whereas protein processing genes were lower expressed in WG than in WS women. CONCLUSION: In adult women, long-term body weight change associates with altered sWAT transcriptome. Expression of genes associated with inflammation, insulin response, adipogenesis and lipogenesis are linked to weight loss. However, other pathways such as autophagy not only associate but also predict future weight gain suggesting that intrinsic factors in sWAT impact tissue expansion.


Subject(s)
Body Weight , Obesity , Subcutaneous Fat, Abdominal/metabolism , Transcriptome/genetics , Adult , Body Weight/genetics , Body Weight/physiology , Female , Humans , Inflammation/genetics , Lipogenesis/genetics , Middle Aged , Obesity/genetics , Obesity/metabolism , Prospective Studies
19.
Int J Obes (Lond) ; 44(2): 544-547, 2020 02.
Article in English | MEDLINE | ID: mdl-31455871

ABSTRACT

Fibroblast growth factor 21 (FGF21) is an important regulator of energy metabolism. FGF21 is inactivated by fibroblast activation protein (FAP). We investigated whether FGF21 and/or FAP are secreted from human white adipose tissue of individuals with obesity by measuring total FGF21, active FGF21, and FAP concentrations in arterialized blood and venous blood draining the subcutaneous abdominal adipose tissue (scAT). Measurements were performed under fasting conditions and after a high fat meal before and after diet-induced weight loss in 16 adults with BMI 27-35 kg/m2. FGF21 was not released from scAT, neither before nor after weight loss in agreement with an undetectable gene expression of FGF21 in this tissue. Although scAT showed significant gene expression of FAP, no release of FAP from the tissue could be detected. The high fat meal increased postprandial circulating FGF21 but not FAP. Circulating levels of FAP but not FGF21 were significantly reduced after weight loss. On the other hand, FAP expression in scAT was increased. In conclusion, release from scAT does not appear to contribute to circulating concentrations of FGF21 and FAP and their responses to ingestion of a high fat meal or weight loss, respectively, in individuals with obesity.


Subject(s)
Fibroblast Growth Factors/blood , Gelatinases/blood , Membrane Proteins/blood , Obesity , Serine Endopeptidases/blood , Subcutaneous Fat, Abdominal/metabolism , Adult , Diet, Reducing , Endopeptidases , Humans , Obesity/blood , Obesity/diet therapy , Obesity/metabolism
20.
Cancer Causes Control ; 31(8): 723-735, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32430684

ABSTRACT

PURPOSE: Underlying mechanisms of the relationship between body fatness and colorectal cancer remain unclear. This study investigated associations of circulating metabolites with visceral (VFA), abdominal subcutaneous (SFA), and total fat area (TFA) in colorectal cancer patients. METHODS: Pre-surgery plasma samples from 212 patients (stage I-IV) from the ColoCare Study were used to perform targeted metabolomics. VFA, SFA, and TFA were quantified by computed tomography scans. Partial correlation and linear regression analyses of VFA, SFA, and TFA with metabolites were computed and corrected for multiple testing. Cox proportional hazards were used to assess 2-year survival. RESULTS: In patients with metastatic tumors, SFA and TFA were statistically significantly inversely associated with 16 glycerophospholipids (SFA: pFDR range 0.017-0.049; TFA: pFDR range 0.029-0.048), while VFA was not. Doubling of ten of the aforementioned glycerophospholipids was associated with increased risk of death in patients with metastatic tumors, but not in patients with non-metastatic tumors (phet range: 0.00044-0.049). Doubling of PC ae C34:0 was associated with ninefold increased risk of death in metastatic tumors (Hazard Ratio [HR], 9.05; 95% confidence interval [CI] 2.17-37.80); an inverse association was observed in non-metastatic tumors (HR 0.17; 95% CI 0.04-0.87; phet = 0.00044). CONCLUSION: These data provide initial evidence that glycerophospholipids in metastatic colorectal cancer are uniquely associated with subcutaneous adiposity, and may impact overall survival.


Subject(s)
Colorectal Neoplasms/metabolism , Intra-Abdominal Fat/metabolism , Subcutaneous Fat, Abdominal/metabolism , Adiposity , Adolescent , Adult , Aged , Aged, 80 and over , Body Mass Index , Colorectal Neoplasms/diagnostic imaging , Colorectal Neoplasms/pathology , Female , Humans , Intra-Abdominal Fat/diagnostic imaging , Male , Metabolomics , Middle Aged , Neoplasm Staging , Subcutaneous Fat, Abdominal/diagnostic imaging , Tomography, X-Ray Computed , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL