Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Diabetes Obes Metab ; 24(5): 908-917, 2022 05.
Article in English | MEDLINE | ID: mdl-35060297

ABSTRACT

AIM: To evaluate the pharmacokinetics and efficacy of a novel somatostatin receptor 2 antagonist, ZT-01, to stimulate glucagon release in rats with type 1 diabetes (T1D). METHODS: The pharmacokinetics of ZT-01 and PRL-2903 were assessed following intraperitoneal or subcutaneous dosing at 10 mg/kg. We compared the efficacy of ZT-01 with PRL-2903 to prevent hypoglycaemia during an insulin bolus challenge and under hypoglycaemic clamp conditions. RESULTS: Within 1 hour after intraperitoneal administration, ZT-01 achieved more than 10-fold higher plasma Cmax compared with PRL-2903. Twenty-four hour exposure was 4.7× and 11.3× higher with ZT-01 by the intraperitoneal and subcutaneous routes, respectively. The median time to reach hypoglycaemia of more than 3.0 mmol/L was 60, 70, and 125 minutes following vehicle, PRL-2903, or ZT-01 administration, respectively. Furthermore, rats receiving ZT-01 had significantly higher glucose nadirs following insulin administration compared with PRL-2903- and vehicle-treated rats. During the hypoglycaemic clamp, ZT-01 increased peak glucagon responses by ~4-fold over PRL-2903. CONCLUSIONS: We conclude that ZT-01 may be effective in restoring glucagon responses and preventing the onset of hypoglycaemia in patients with T1D.


Subject(s)
Diabetes Mellitus, Type 1 , Hypoglycemia , Receptors, Somatostatin , Animals , Blood Glucose , Diabetes Mellitus, Type 1/drug therapy , Glucagon , Humans , Hypoglycemia/chemically induced , Hypoglycemia/prevention & control , Insulin , Rats , Receptors, Somatostatin/antagonists & inhibitors
2.
Gastroenterology ; 156(8): 2297-2312, 2019 06.
Article in English | MEDLINE | ID: mdl-30836096

ABSTRACT

BACKGROUND & AIMS: Interleukin 6 (IL6) and tumor necrosis factor contribute to the development of colitis-associated cancer (CAC). We investigated these signaling pathways and the involvement of G protein subunit alpha i1 (GNAI1), GNAI2, and GNAI3 in the development of CAC in mice and humans. METHODS: B6;129 wild-type (control) or mice with disruption of Gnai1, Gnai2, and/or Gnai3 or conditional disruption of Gnai2 in CD11c+ or epithelial cells were given dextran sulfate sodium (DSS) to induce colitis followed by azoxymethane (AOM) to induce carcinogenesis; some mice were given an antibody against IL6. Feces were collected from mice, and the compositions of microbiomes were analyzed by polymerase chain reactions. Dendritic cells (DCs) and myeloid-derived suppressor cells (MDSCs) isolated from spleen and colon tissues were analyzed by flow cytometry. We performed immunoprecipitation and immunoblot analyses of colon tumor tissues, MDSCs, and mouse embryonic fibroblasts to study the expression levels of GNAI1, GNAI2, and GNAI3 and the interactions of GNAI1 and GNAI3 with proteins in the IL6 signaling pathway. We analyzed the expression of Gnai2 messenger RNA by CD11c+ cells in the colonic lamina propria by PrimeFlow, expression of IL6 in DCs by flow cytometry, and secretion of cytokines in sera and colon tissues by enzyme-linked immunosorbent assay. We obtained colon tumor and matched nontumor tissues from 83 patients with colorectal cancer having surgery in China and 35 patients with CAC in the United States. Mouse and human colon tissues were analyzed by histology, immunoblot, immunohistochemistry, and/or RNA-sequencing analyses. RESULTS: GNAI1 and GNAI3 (GNAI1;3) double-knockout (DKO) mice developed more severe colitis after administration of DSS and significantly more colonic tumors than control mice after administration of AOM plus DSS. Development of increased tumors in DKO mice was not associated with changes in fecal microbiomes but was associated with activation of nuclear factor (NF) κB and signal transducer and activator of transcription (STAT) 3; increased levels of GNAI2, nitric oxide synthase 2, and IL6; increased numbers of CD4+ DCs and MDSCs; and decreased numbers of CD8+ DCs. IL6 was mainly produced by CD4+/CD11b+, but not CD8+, DCs in DKO mice. Injection of DKO mice with a blocking antibody against IL6 reduced the expansion of MDSCs and the number of tumors that developed after CAC induction. Incubation of MDSCs or mouse embryonic fibroblasts with IL6 induced activation of either NF-κB by a JAK2-TRAF6-TAK1-CHUK/IKKB signaling pathway or STAT3 by JAK2. This activation resulted in expression of GNAI2, IL6 signal transducer (IL6ST, also called GP130) and nitric oxide synthase 2, and expansion of MDSCs; the expression levels of these proteins and expansion of MDSCs were further increased by the absence of GNAI1;3 in cells and mice. Conditional disruption of Gnai2 in CD11c+ cells of DKO mice prevented activation of NF-κB and STAT3 and changes in numbers of DCs and MDSCs. Colon tumor tissues from patients with CAC had reduced levels of GNAI1 and GNAI3 and increased levels of GNAI2 compared with normal tissues. Further analysis of a public human colorectal tumor DNA microarray database (GSE39582) showed that low Gani1 and Gnai3 messenger RNA expression and high Gnai2 messenger RNA expression were significantly associated with decreased relapse-free survival. CONCLUSIONS: GNAI1;3 suppresses DSS-plus-AOM-induced colon tumor development in mice, whereas expression of GNAI2 in CD11c+ cells and IL6 in CD4+/CD11b+ DCs appears to promote these effects. Strategies to induce GNAI1;3, or block GNAI2 and IL6, might be developed for the prevention or therapy of CAC in patients.


Subject(s)
Cell Transformation, Neoplastic/genetics , Colitis/pathology , Colonic Neoplasms/pathology , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Animals , Biopsy, Needle , Carcinogenesis , Colitis/genetics , Colonic Neoplasms/genetics , Disease Models, Animal , Down-Regulation/genetics , Female , Gene Expression Regulation, Neoplastic , Immunohistochemistry , Interleukin-16/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Random Allocation , Reference Values , Sensitivity and Specificity , Signal Transduction/genetics
3.
J Transl Med ; 18(1): 57, 2020 02 05.
Article in English | MEDLINE | ID: mdl-32024545

ABSTRACT

BACKGROUND: Accumulating evidence suggests that plasminogen activator inhibitor-1 (PAI-1) plays an important role in bladder tumorigenesis by regulating cell cycle. However, it remains unclear whether and how inhibition of PAI-1 suppresses bladder tumorigenesis. METHODS: To elucidate the therapeutic effect of PAI-1 inhibition, we tested its tumorigenicity in PAI-1 knockout (KO) mice exposed to a known bladder carcinogen. RESULTS: PAI-1 deficiency did not inhibit carcinogen-induced bladder cancer in mice although carcinogen-exposed wild type mice significantly increased PAI-1 levels in bladder tissue, plasma and urine. We found that PAI-1 KO mice exposed to carcinogen tended to upregulate protein C inhibitor (PAI-3), urokinase-type plasminogen activator (uPA) and tissue-type PA (tPA), and significantly increased PAI-2, suggesting a potential compensatory function of these molecules when PAI-1 is abrogated. Subsequent studies employing gene expression microarray using mouse bladder tissues followed by post hoc bioinformatics analysis and validation experiments by qPCR and IHC demonstrated that SERPING1 is further downregulated in PAI-1 KO mice exposed to BBN, suggesting that SERPING1 as a potential missing factor that regulate PAI-2 overexpression (compensation pathway). CONCLUSIONS: These results indicate that serpin compensation pathway, specifically PAI-2 overexpression in this model, supports bladder cancer development when oncoprotein PAI-1 is deleted. Further investigations into PAI-1 are necessary in order to identify true potential targets for bladder cancer therapy.


Subject(s)
Plasminogen Activator Inhibitor 1 , Plasminogen Activator Inhibitor 2 , Urinary Bladder Neoplasms , Animals , Mice , Mice, Knockout , Nitrosamines , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 2/genetics , Serpin E2 , Urinary Bladder Neoplasms/chemically induced , Urinary Bladder Neoplasms/genetics
4.
Diabetologia ; 62(4): 676-686, 2019 04.
Article in English | MEDLINE | ID: mdl-30627753

ABSTRACT

AIMS/HYPOTHESIS: This study evaluates whether the non-selective ß-blocker, carvedilol, can be used to prevent counterregulatory failure and the development of impaired awareness of hypoglycaemia (IAH) in recurrently hypoglycaemic rats. METHODS: Sprague Dawley rats were implanted with vascular catheters and intracranial guide cannulas targeting the ventromedial hypothalamus (VMH). These animals underwent either three bouts of insulin-induced hypoglycaemia or received three saline injections (control group) over 3 days. A subgroup of recurrently hypoglycaemic animals was treated with carvedilol. The next day, the animals underwent a hypoglycaemic clamp with microdialysis without carvedilol treatment to evaluate changes in central lactate and hormone levels. To assess whether carvedilol prevented IAH, we treated rats that had received repeated 2-deoxyglucose (2DG) injections to impair their awareness of hypoglycaemia with carvedilol and measured food intake in response to insulin-induced hypoglycaemia as a surrogate marker for hypoglycaemia awareness. RESULTS: Compared with the control group, recurrently hypoglycaemic rats had a ~1.7-fold increase in VMH lactate and this was associated with a 75% reduction in the sympathoadrenal response to hypoglycaemia. Treatment with carvedilol restored VMH lactate levels and improved the adrenaline (epinephrine) responses. In 2DG-treated rats compared with control animals receiving saline, food intake was reduced in response to hypoglycaemia and increased with carvedilol treatment. CONCLUSIONS/INTERPRETATION: We conclude that carvedilol may be a useful therapy to prevent counterregulatory failure and improve IAH.


Subject(s)
Adrenergic beta-1 Receptor Antagonists/pharmacology , Carvedilol/therapeutic use , Hyperglycemia/drug therapy , Hypoglycemia/drug therapy , Hypoglycemia/prevention & control , Animals , Blood Glucose , Body Weight , Catheterization , Deoxyglucose/administration & dosage , Disease Models, Animal , Glucose Clamp Technique , Hypoglycemic Agents/adverse effects , Insulin/adverse effects , Lactic Acid/blood , Male , Rats , Rats, Sprague-Dawley , Recurrence , Time Factors , Ventromedial Hypothalamic Nucleus/drug effects
5.
Am J Physiol Endocrinol Metab ; 316(5): E948-E955, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30888861

ABSTRACT

Increased GABAergic output in the ventromedial hypothalamus (VMH) contributes to counterregulatory failure in recurrently hypoglycemic (RH) rats, and lactate, an alternate fuel source in the brain, contributes to this phenomenon. The current study assessed whether recurring bouts of glucose deprivation enhanced neuronal lactate uptake and, if so, whether this influenced γ-aminobutyric acid (GABA) output and the counterregulatory responses. Glucose deprivation was induced using 5-thioglucose (5TG). Control rats received an infusion of artificial extracellular fluid. These groups were compared with RH animals. Subsequently, the rats underwent a hypoglycemic clamp with microdialysis. To test whether 5TG affected neuronal lactate utilization, a subgroup of 5TG-treated rats was microinjected with a lactate transporter inhibitor [cyano-4-hydroxycinnamate (4CIN)] just before the start of the clamp. Both RH and 5TG raised VMH GABA levels, and this was associated with impaired counterregulatory responses. 4CIN reduced VMH GABA levels and restored the hormone responses in the 5TG group. We then evaluated [14C]lactate uptake in hypothalamic neuronal cultures. Recurring exposure to low glucose increased monocarboxylate transporter-2 mRNA expression and augmented lactate uptake. Taken together, our data suggest that glucose deprivation, per se, enhances lactate utilization in hypothalamic neurons, and this may contribute to suppression of the counterregulatory responses to hypoglycemia.


Subject(s)
Glucose/metabolism , Hypoglycemia/metabolism , Hypothalamus, Middle/cytology , Lactic Acid/metabolism , Neurons/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Carbon Radioisotopes , Catecholamines/metabolism , Coumaric Acids/pharmacology , Glucose/analogs & derivatives , Glucose/deficiency , Glucose/pharmacology , Glucose Clamp Technique , Hypothalamus, Middle/drug effects , Hypothalamus, Middle/metabolism , Microdialysis , Monocarboxylic Acid Transporters/antagonists & inhibitors , Monocarboxylic Acid Transporters/drug effects , Monocarboxylic Acid Transporters/genetics , Neurons/drug effects , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , gamma-Aminobutyric Acid/drug effects
6.
Clin Gastroenterol Hepatol ; 17(10): 2129-2131, 2019 09.
Article in English | MEDLINE | ID: mdl-30448596

ABSTRACT

Rectal squamous cell carcinoma (SCC) is a rare tumor with unresolved etiology. Human immunodeficiency virus-infected individuals and solid organ transplant recipients experience >30-fold and approximately 3-fold elevated rates of rectal SCC, respectively, suggesting immunosuppression plays a role.1 Human immunodeficiency virus-infected homosexual men have >60-fold higher rates of rectal SCC, similar to anal SCC. These patterns, which differ from the more common rectal adenocarcinoma (AdCA), raise the possibility of shared etiology between rectal and anal SCC, with human papillomavirus type 16 (HPV16) being a likely candidate.2.


Subject(s)
Carcinoma, Squamous Cell/pathology , Human papillomavirus 16/genetics , Papillomavirus Infections/epidemiology , Rectal Neoplasms/pathology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma/virology , Anus Neoplasms/metabolism , Anus Neoplasms/pathology , Anus Neoplasms/virology , Biomarkers/metabolism , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/virology , Case-Control Studies , DNA, Viral/analysis , DNA-Binding Proteins/genetics , Humans , In Situ Hybridization , Keratins/metabolism , Oncogene Proteins, Viral/genetics , Papillomavirus Infections/virology , Rectal Neoplasms/metabolism , Rectal Neoplasms/virology , Repressor Proteins/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Viral Envelope Proteins/genetics
7.
J Transl Med ; 17(1): 29, 2019 01 17.
Article in English | MEDLINE | ID: mdl-30654801

ABSTRACT

BACKGROUND: We set out to determine if the administration of subcutaneous (SQ) ALT-803 was non-inferior to standard intravesical BCG treatment in a carcinogen induced mouse (C57BL/6J) bladder cancer model. METHODS: Using this well-established carcinogen induced mouse model, we studied the effects of various dosing schemas of ALT-803 (SQ alone, SQ with intravesical BCG, intravesical alone, intravesical with intravesical BCG) compared to intravesical BCG alone (positive control) and PBS (negative control). The non-inferiority margin for the difference in bladder weight, as a surrogate for tumor mass, was defined as 7%. RESULTS: All treatment groups (i.e., ALT-803 SQ alone, ALT-803 SQ with intravesical BCG, ALT-803 intravesical alone, ALT-803 intravesical with intravesical BCG and intravesical BCG alone) demonstrated a significant reduction in tumor burden as evident by bladder weights and H&E stain (p < 0.005). Non-inferiority tests between the intravesical BCG alone group and the additional treatment groups showed that SQ ALT-803 alone (p = 0.04) and BCG plus SQ ALT-803 (p = 0.009) were non-inferior to intravesical BCG alone. In this model, we did not see an appreciable infiltration of CD4+ T, CD8+ T or CD161/KLRB1+ natural killer (NK) cells in the bladder/tumor. When assessing peripheral blood mononuclear cells, SQ ALT-803 alone resulted in a robust induction of CD8+ T cells (p < 0.01), NKG2D+ NK cells (p < 0.005) and CD3+/NKG2D+ NKT cells (p < 0.005) compared to other groups, while in splenic tissue, SQ ALT-803 alone resulted in a robust induction of CD3+/NKG2D+ NKT cells (p < 0.005) compared to other groups. CONCLUSION: Subcutaneous ALT-803 treatment alone or in combination with intravesical BCG was well tolerated and was not inferior to intravesical BCG alone. CD8+ T, NKG2D+ NK and CD3+/NKG2D+ NKT cell induction along with induction of key cytokines remain steadfast mechanisms behind ALT-803. The enhanced therapeutic index seen with BCG and ALT-803, administered SQ or intravesically, provides a powerful justification for the further development of these regimens.


Subject(s)
Interleukin-15/agonists , Proteins/administration & dosage , Proteins/therapeutic use , Urinary Bladder Neoplasms/drug therapy , Animals , Cytokines/blood , Cytokines/urine , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Therapy, Combination , Female , Interleukin-15/metabolism , Lymphocytes/metabolism , Mice, Inbred C57BL , Mycobacterium bovis , Proteins/pharmacology , Recombinant Fusion Proteins , Treatment Outcome , Urinary Bladder Neoplasms/blood , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/urine
8.
Endocr Pract ; 25(6): 517-525, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30865520

ABSTRACT

Objective: Impaired awareness of hypoglycemia (IAH) is a risk factor for severe hypoglycemia in patients with type 1 diabetes (T1D) not using a continuous glucose monitoring (CGM) system. The current study investigated the prevalence of IAH and its relationship with severe hypoglycemia in T1D patients using CGM systems. Methods: This cross-sectional observational study enrolled 135 patients with T1D and ongoing real-time CGM use. A survey was conducted to assess hypoglycemia awareness with the Gold, Clarke, and Pedersen-Bjergaard questionnaires and the 6-month history of severe hypoglycemia. Other diabetes histories and the CGM glucose data were collected. Results: The Gold, Clarke, and Pedersen-Bjergaard questionnaires demonstrated the overall prevalence of IAH/abnormal awareness to be 33.3%, 43.7%, and 77.0%, respectively. Participant age and duration of T1D were consistently related to IAH or hypoglycemia unawareness with all three questionnaires (P<.05). Amongst the patients using CGM for >6 months, 24.5% were found to have at least one episode of severe hypoglycemia in the preceding 6 months. IAH identified by the Gold and Clarke questionnaires and hypoglycemia unawareness identified by the Pedersen-Bjergaard questionnaire were related to 6-, 4.63-, and 5.83-fold increased risk of severe hypoglycemia (P = .001, .004, and .013), respectively. IAH identified by the Gold/Clarke questionnaires was associated with a longer duration of CGM glucose <54 mg/dL and higher glucose coefficients of variation (P<.05). Conclusion: IAH is highly prevalent and related to a higher risk for severe hypoglycemia in T1D patients using CGM. Abbreviations: CGM = continuous glucose monitoring; CI = confidence interval; HAAF = hypoglycemia-associated autonomic failure; HbA1c = hemoglobin A1C; IAH = impaired awareness of hypoglycemia; T1D = type 1 diabetes.


Subject(s)
Diabetes Mellitus, Type 1 , Hypoglycemia , Blood Glucose , Blood Glucose Self-Monitoring , Cross-Sectional Studies , Glycated Hemoglobin , Humans , Risk Factors
9.
Carcinogenesis ; 39(1): 47-55, 2018 01 12.
Article in English | MEDLINE | ID: mdl-28968647

ABSTRACT

Aberrant sphingolipid metabolism has been reported to promote breast cancer progression. Sphingosine kinase 1 (SphK1) is a key metabolic enzyme for the formation of pro-survival S1P from pro-apoptotic ceramide. The role of SphK1 in breast cancer has been well studied in estrogen receptor (ER)-positive breast cancer; however, its role in human epidermal growth factor 2 (HER2)-positive breast cancer remains unclear. Here, we show that genetic deletion of SphK1 significantly reduced mammary tumor development with reduced tumor incidence and multiplicity in the MMTV-neu transgenic mouse model. Gene expression analysis revealed significant reduction of claudin-2 (CLDN2) expression in tumors from SphK1 deficient mice, suggesting that CLDN2 may mediate SphK1's function. It is remarkable that SphK1 deficiency in HER2-positive breast cancer model inhibited tumor formation by the different mechanism from ER-positive breast cancer. In vitro experiments demonstrated that overexpression of SphK1 in ER-/PR-/HER2+ human breast cancer cells enhanced cell proliferation, colony formation, migration and invasion. Furthermore, immunostaining of SphK1 and CLDN2 in HER2-positive human breast tumors revealed a correlation in high-grade disease. Taken together, these findings suggest that SphK1 may play a pivotal role in HER2-positive breast carcinogenesis. Targeting SphK1 may represent a novel approach for HER2-positive breast cancer chemoprevention and/or treatment.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Phosphotransferases (Alcohol Group Acceptor)/genetics , Receptor, ErbB-2/genetics , Animals , Breast Neoplasms/metabolism , Disease Models, Animal , Female , Humans , Mice , Mice, Transgenic
10.
Am J Physiol Endocrinol Metab ; 315(6): E1286-E1295, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30226996

ABSTRACT

It is proposed that the impaired counterregulatory response (CRR) to hypoglycemia in insulin-deficient diabetes may be due to chronic brain insulin deficiency. To test this hypothesis, streptozotocin-induced diabetic Sprague-Dawley rats were infused with insulin (3 mU/day) or artificial cerebrospinal fluid (aCSF) bilaterally into the ventromedial hypothalamus (VMH) for 2 wk and compared with nondiabetic rats. Rats underwent hyperinsulinemic (50 mU·kg-1·min-1)-hypoglycemic (~45 mg/dl) clamps. Diabetic rats demonstrated an impaired CRR to hypoglycemia, noted by a high glucose infusion rate and blunted epinephrine and glucagon responses. The defective sympathoadrenal response was restored by chronic infusion of insulin into the VMH. Diabetic rats had decreased VMH Akt phosphorylation and decreased VMH glucose transporter 4 (GLUT4) content, which was also restored by chronic infusion of insulin into the VMH. Separate experiments in nondiabetic rats in which GLUT4 translocation into the VMH was inhibited with an infusion of indinavir were notable for an impaired CRR to hypoglycemia, indicated by increased glucose infusion rate and diminished epinephrine and glucagon responses. Results suggest that, in this model of diabetes, VMH insulin deficiency impairs the sympathoadrenal response to hypoglycemia and that chronic infusion of insulin into the VMH is sufficient to normalize the sympathoadrenal response to hypoglycemia via restoration of GLUT4 expression in the VMH.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Experimental/metabolism , Glucose Transporter Type 4/metabolism , Hypoglycemia/metabolism , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Ventromedial Hypothalamic Nucleus/drug effects , Animals , Epinephrine/blood , Glucagon/blood , Glucose Clamp Technique , Male , Rats , Rats, Sprague-Dawley , Ventromedial Hypothalamic Nucleus/metabolism
11.
Radiology ; 287(1): 294-302, 2018 04.
Article in English | MEDLINE | ID: mdl-29315063

ABSTRACT

Purpose To determine the relationship between hepatic uptake at preoperative fluorine 18 (18F) fluorocholine combined positron emission tomography (PET) and computed tomography (CT) and the histopathologic features of chronic liver disease in patients with Child-Pugh class A or B disease who are undergoing hepatic resection for liver cancer. Materials and Methods Forty-eight patients with resectable liver tumors underwent preoperative 18F fluorocholine PET/CT. Mean liver standardized uptake value (SUVmean) measurements were obtained from PET images, while histologic indexes of inflammation and fibrosis were applied to nontumor liver tissue from resection specimens. Effects of histopathologic features on liver SUVmean were examined with analysis of variance. Results Liver SUVmean ranged from 4.3 to 11.6, correlating significantly with Knodell histologic activity index (ρ = -0.81, P < .001) and several clinical indexes of liver disease severity. Liver SUVmean also differed significantly across groups stratified by necroinflammatory severity and Metavir fibrosis stage (P < . 001). The area under the receiver operating characteristic curve for 18F fluorocholine PET/CT detecting Metavir fibrosis stage F1 or higher was 0.89 ± 0.05, with an odds-ratio of 3.03 (95% confidence interval: 1.59, 5.88) and sensitivity and specificity of 82% and 93%, respectively. Conclusion Correlations found in patients undergoing hepatic resection for liver cancer between liver 18F fluorocholine uptake and histopathologic indexes of liver fibrosis and inflammation support the use of 18F fluorocholine PET/CT as a potential imaging biomarker for chronic liver disease. © RSNA, 2018.


Subject(s)
Choline/analogs & derivatives , Liver Neoplasms/diagnostic imaging , Positron Emission Tomography Computed Tomography/methods , Radiopharmaceuticals , Aged , Biomarkers , Chronic Disease , Female , Fluorine Radioisotopes , Humans , Liver/diagnostic imaging , Male , Middle Aged , Prospective Studies , Reproducibility of Results , Sensitivity and Specificity
12.
Int J Exp Pathol ; 99(4): 172-179, 2018 08.
Article in English | MEDLINE | ID: mdl-30256482

ABSTRACT

This study was designed to investigate the HPA-axis impairment in the streptozotocin (STZ)-diabetic gerbils (Gerbillus gerbillus). Twenty-six male gerbils (body weight ~27 g) were divided into 3 groups: vehicle control (n = 10), 2 days of diabetes (n = 09) and 30 days of diabetes (n = 07). The latter 2 groups received an intraperitoneal injection of STZ (150 mg/kg of body weight). At 2 and 30 days of diabetes, streptozotocin-diabetic gerbils underwent a retro-orbital puncture for assessment of biochemical and hormonal parameters. Subsequently the animals were decapitated and the adrenal glands were removed, weighed and processed for light microscopy and stereology. Nondiabetic control gerbils that had been injected with citrate buffer were examined as a comparison. At 2 days of diabetes, STZ gerbils exhibited symptoms that are characteristic of human diabetes type 1. The adrenal gland showed significant increase in weight, associated with a larger cortex layer, hypertrophy of the fasciculate cells and a significant decrease in the nucleocytoplasmic index. These changes were associated with higher plasma ACTH and cortisol concentrations compared to nondiabetic controls. At 30 days postdiabetes, ACTH levels remained elevated, whereas cortisol levels decreased compared to the early stage of diabetes. Histological analysis revealed the existence of a band of connective tissue (collagen) that separates the cortical and medullary zones and is not present in humans or laboratory rodents, which represents a striking change seen throughout the disease. STZ-induced diabetes mellitus in Gerbillus gerbillus resulted in hyperactivation of the HPA axis in the early stages of diabetes mellitus which did not persist into the final stages of the disease, suggesting a possible reduction in adrenocortical sensitivity over time.


Subject(s)
Diabetes Mellitus, Type 1/pathology , Hypothalamo-Hypophyseal System/pathology , Pituitary-Adrenal System/pathology , Streptozocin/pharmacology , Animals , Body Weight/physiology , Corticosterone/blood , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Type 1/chemically induced , Gerbillinae/physiology , Insulin/metabolism , Male
13.
BMC Cancer ; 18(1): 1191, 2018 Nov 29.
Article in English | MEDLINE | ID: mdl-30497427

ABSTRACT

BACKGROUND: Mammographic density is a known risk factor for breast cancer, but the underlying pathologic characteristics are not well understood. The current analysis investigated the expression of several markers of interest, e.g., inflammation and growth, with mammographic density (MD) in normal and malignant breast tissue specimens from 279 women of the Multiethnic Cohort (MEC). METHODS: Breast cancer cases, recruited from a nested case-control study within the MEC, provided mammograms for density evaluation. Protein expression (COX-2, TNF-α, TGF-ß, IGF-1R, IGFBP-2, and vimentin) was assessed by immunohistochemical detection. Linear regression was applied to evaluate the relation between marker expression and percent density and to compute adjusted means with 95% confidence intervals (CI) by marker status while adjusting for confounders. RESULTS: Due to missing cores and tissue, normal tissue could only be evaluated for COX-2 and vimentin. No significant associations with mammographic density were detected for all markers analyzed. For inflammatory markers (TNF-α, COX-2, and TGF-ß) in tumor tissue, MD were non-significantly higher with stronger expression but the differences were very small. For example, the mean MD values for no, weak, and strong TNF-α expression were 35% (95% CI 24-47%), 39% (95% CI 29-48%), and 38% (95% CI 27-50%). In a posthoc analysis among postmenopausal women only, the difference across categories of TNF-α expression increased to 25% (95% CI 12-39%), 35% (95% CI 23-48%), and 35% (95% CI 20-49%). CONCLUSIONS: The current analysis offers little support for an involvement of immunohistochemical markers representing inflammatory and growth factor pathways as predictors of breast density.


Subject(s)
Breast Density , Gene Expression , Inflammation Mediators/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Mammary Glands, Human/diagnostic imaging , Mammary Glands, Human/metabolism , Vimentin/metabolism , Adult , Aged , Biomarkers , Case-Control Studies , Female , Humans , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/genetics , Mammography , Middle Aged , Vimentin/genetics
14.
J Transl Med ; 15(1): 120, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28583134

ABSTRACT

BACKGROUND: Accumulating evidence suggests that sphingosine kinase 1 (SphK1)/sphingosine 1-phosphate pathway plays a pivotal role in colon carcinogenesis. METHODS: To further support the evidence, we investigated the effects of SphK1 using three separate animal models: SphK1 knockout mice, SphK1 overexpressing transgenic mice, and SphK1 overexpression in human colon cancer xenografts. Using azoxymethane (AOM, colon carcinogen), we analyzed colon tumor development in SphK1 KO and SphK1 overexpression in intestinal epithelial cells regulated by a tet-on system. Then, we analyzed subcutaneous tumor growth using xenografts of HT-29 human colon cancer cell. Finally, immunohistochemical analyses for SphK1 and COX-2 were performed on human colon cancer tissue microarray. RESULTS: SphK1 KO mice, compared to wild-type mice, demonstrated a significant inhibition in colon cancer development induced by AOM (58.6% vs. 96.4%, respectively, P < 0.005). Tumor multiplicity (1.00 vs. 1.64 per colon, respectively, P < 0.05) and tumor volume (14.82 mm3 vs. 29.10 mm3, P < 0.05) were both significantly reduced in SphK1 KO mice compared to wild-type mice. Next, SphK1 overexpression in HT-29 enhanced tumor growth as compared to GFP control in nude mice (229.5 mm3 vs. 90.9 mm3, respectively, P < 0.05). Furthermore, overexpression of SphK1 in intestinal epithelial cells significantly enhances AOM-induced colon tumor formation (P < 0.05). Lastly, SphK1 and COX-2 intensity tended to reduce overall survival of late stage colon cancer patients. CONCLUSIONS: SphK1 expression regulates the early stage of colon carcinogenesis and tumor growth, thus inhibition of SphK1 may be an effective strategy for colon cancer chemoprevention.


Subject(s)
Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Aged , Animals , Azoxymethane , Carcinogenesis/pathology , Cell Proliferation , Cyclooxygenase 2/metabolism , Enterocytes/metabolism , Enterocytes/pathology , Female , HT29 Cells , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Neoplasm Staging
15.
Proc Natl Acad Sci U S A ; 111(32): 11876-81, 2014 Aug 12.
Article in English | MEDLINE | ID: mdl-25071172

ABSTRACT

Prolyl endopeptidase (PREP) has been implicated in neuronal functions. Here we report that hypothalamic PREP is predominantly expressed in the ventromedial nucleus (VMH), where it regulates glucose-induced neuronal activation. PREP knockdown mice (Prep(gt/gt)) exhibited glucose intolerance, decreased fasting insulin, increased fasting glucagon levels, and reduced glucose-induced insulin secretion compared with wild-type controls. Consistent with this, central infusion of a specific PREP inhibitor, S17092, impaired glucose tolerance and decreased insulin levels in wild-type mice. Arguing further for a central mode of action of PREP, isolated pancreatic islets showed no difference in glucose-induced insulin release between Prep(gt/gt) and wild-type mice. Furthermore, hyperinsulinemic euglycemic clamp studies showed no difference between Prep(gt/gt) and wild-type control mice. Central PREP regulation of insulin and glucagon secretion appears to be mediated by the autonomic nervous system because Prep(gt/gt) mice have elevated sympathetic outflow and norepinephrine levels in the pancreas, and propranolol treatment reversed glucose intolerance in these mice. Finally, re-expression of PREP by bilateral VMH injection of adeno-associated virus-PREP reversed the glucose-intolerant phenotype of the Prep(gt/gt) mice. Taken together, our results unmask a previously unknown player in central regulation of glucose metabolism and pancreatic function.


Subject(s)
Glucagon/metabolism , Hypothalamus/enzymology , Insulin/metabolism , Serine Endopeptidases/metabolism , Animals , Blood Glucose/metabolism , Gene Expression , Gene Knockdown Techniques , Glucose Clamp Technique , Glucose Intolerance/enzymology , Glucose Intolerance/etiology , Hypothalamus/physiology , Indoles/pharmacology , Insulin Secretion , Ion Channels/genetics , Male , Mice , Mice, Transgenic , Mitochondrial Proteins/genetics , Pancreas/metabolism , Phosphorylation , Prolyl Oligopeptidases , Receptor, Insulin/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serine Endopeptidases/deficiency , Serine Endopeptidases/genetics , Serine Proteinase Inhibitors/pharmacology , Thiazolidines/pharmacology , Uncoupling Protein 1 , Ventromedial Hypothalamic Nucleus/enzymology , Ventromedial Hypothalamic Nucleus/physiology
16.
J Transl Med ; 14: 31, 2016 Jan 30.
Article in English | MEDLINE | ID: mdl-26830497

ABSTRACT

BACKGROUND: Urine based assays that can non-invasively detect bladder cancer (BCa) have the potential to reduce unnecessary and invasive procedures. The purpose of this study was to develop a multiplex immunoassay that can accurately and simultaneously monitor ten diagnostic urinary protein biomarkers for application as a non-invasive test for BCa detection. METHODS: A custom electrochemiluminescent multiplex assay was constructed (Meso Scale Diagnostics, LLC, Rockville, MD, USA) to detect the following urinary proteins; IL8, MMP9, MMP10, ANG, APOE, SDC1, A1AT, PAI1, CA9 and VEGFA. Voided urine samples from two cohorts were collected prior to cystoscopy and samples were analyzed blinded to the clinical status of the participants. Means (±SD) and receiver operating characteristic (ROC) curve analysis were used to compare assay performance and to assess the diagnostic accuracy of the diagnostic signature. RESULTS: Comparative diagnostic performance analyses revealed an AUROC value of 0.9258 for the multiplex assay and 0.9467 for the combination of the single-target ELISA assays (p = 0.625), so there was no loss of diagnostic utility for the MSD multiplex assay. Analysis of the independent 200-sample cohort using the multiplex assay achieved an overall diagnostic sensitivity of 0.85, specificity of 0.81, positive predictive value 0.82 and negative predictive value 0.84. CONCLUSIONS: It is technically feasible to simultaneously monitor complex urinary diagnostic signatures in a single assay without loss of performance. The described protein-based assay has the potential to be developed for the non-invasive detection of BCa.


Subject(s)
Immunoassay/methods , Urinary Bladder Neoplasms/diagnosis , Urinary Bladder Neoplasms/pathology , Aged , Aged, 80 and over , Biomarkers, Tumor/urine , Cohort Studies , Demography , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Middle Aged , Neoplasm Invasiveness , Reproducibility of Results , Urinary Bladder Neoplasms/urine
17.
Am J Physiol Endocrinol Metab ; 307(7): E580-8, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25117409

ABSTRACT

Recurrent exposure to hypoglycemia can impair the normal counterregulatory hormonal responses that guard against hypoglycemia, leading to hypoglycemia unawareness. This pathological condition known as hypoglycemia-associated autonomic failure (HAAF) is the main adverse consequence that prevents individuals with type 1 diabetes mellitus from attaining the long-term health benefits of tight glycemic control. The underlying molecular mechanisms responsible for the progressive loss of the epinephrine response to subsequent bouts of hypoglycemia, a hallmark sign of HAAF, are largely unknown. Normally, hypoglycemia triggers both the release and biosynthesis of epinephrine through activation of nicotinic acetylcholine receptors (nAChR) on the adrenal glands. We hypothesize that excessive cholinergic stimulation may contribute to impaired counterregulation. Here, we tested whether administration of the nAChR partial agonist cytisine to reduce postganglionic synaptic activity can preserve the counterregulatory hormone responses in an animal model of HAAF. Compared with nicotine, cytisine has limited efficacy to activate nAChRs and stimulate epinephrine release and synthesis. We evaluated adrenal catecholamine production and secretion in nondiabetic rats subjected to two daily episodes of hypoglycemia for 3 days, followed by a hyperinsulinemic hypoglycemic clamp on day 4. Recurrent hypoglycemia decreased epinephrine responses, and this was associated with suppressed TH mRNA induction (a measure of adrenal catecholamine synthetic capacity). Treatment with cytisine improved glucagon responses as well as epinephrine release and production in recurrently hypoglycemic animals. These data suggest that pharmacological manipulation of ganglionic nAChRs may be promising as a translational adjunctive therapy to avoid HAAF in type 1 diabetes mellitus.


Subject(s)
Adrenal Glands/drug effects , Alkaloids/pharmacology , Autonomic Nervous System/drug effects , Blood Glucose/drug effects , Epinephrine/metabolism , Hypoglycemia/metabolism , Nicotinic Antagonists/pharmacology , Receptors, Nicotinic , Tyrosine 3-Monooxygenase/drug effects , Adrenal Glands/metabolism , Animals , Autonomic Nervous System/metabolism , Azocines/pharmacology , Blood Glucose/metabolism , Disease Models, Animal , Epinephrine/biosynthesis , Gene Expression Profiling , Glucose Clamp Technique , Male , Quinolizines/pharmacology , RNA, Messenger , Rats , Rats, Sprague-Dawley , Recurrence , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism
19.
Case Reports Plast Surg Hand Surg ; 11(1): 2400138, 2024.
Article in English | MEDLINE | ID: mdl-39262928

ABSTRACT

We describe the first examples of breast capsular contracture amelioration using a non-surgical, transdermal treatment with platelet-rich plasma. The treated patients did not experience any complications or significant pain. This report illustrates the potential of a non-invasive treatment option for a common complication of breast augmentation.

20.
Front Pharmacol ; 15: 1302015, 2024.
Article in English | MEDLINE | ID: mdl-38510652

ABSTRACT

Background: Elevated levels of somatostatin blunt glucagon counterregulation during hypoglycemia in type 1 diabetes (T1D) and this can be improved using somatostatin receptor 2 (SSTR2) antagonists. Hypoglycemia also occurs in late-stage type 2 diabetes (T2D), particularly when insulin therapy is initiated, but the utility of SSTR2 antagonists in ameliorating hypoglycemia in this disease state is unknown. We examined the efficacy of a single-dose of SSTR2 antagonists in a rodent model of T2D. Methods: High-fat fed (HFF), low dose streptozotocin (STZ, 35 mg/kg)-induced T2D and HFF only, nondiabetic (controls-no STZ) rats were treated with the SSTR2 antagonists ZT-01/PRL-2903 or vehicle (n = 9-11/group) 60 min before an insulin tolerance test (ITT; 2-12 U/kg insulin aspart) or an oral glucose tolerance test (OGTT; 2 g/kg glucose via oral gavage) on separate days. Results: This rodent model of T2D is characterized by higher baseline glucose and HbA1c levels relative to HFF controls. T2D rats also had lower c-peptide levels at baseline and a blunted glucagon counterregulatory response to hypoglycemia when subjected to the ITT. SSTR2 antagonists increased the glucagon response and reduced incidence of hypoglycemia, which was more pronounced with ZT-01 than PRL-2903. ZT-01 treatment in the T2D rats increased glucagon levels above the control response within 60 min of dosing, and values remained elevated during the ITT (glucagon Cmax: 156 ± 50 vs. 77 ± 46 pg/mL, p < 0.01). Hypoglycemia incidence was attenuated with ZT-01 vs. controls (63% vs. 100%) and average time to hypoglycemia onset was also delayed (103.1 ± 24.6 vs. 66.1 ± 23.6 min, p < 0.05). ZT-01 administration at the OGTT onset increased the glucagon response without exacerbating hyperglycemia (2877 ± 806 vs. 2982 ± 781), potentially due to the corresponding increase in c-peptide levels (6251 ± 5463 vs. 14008 ± 5495, p = 0.013). Conclusion: Treatment with SSTR2 antagonists increases glucagon responses in a rat model of T2D and results in less hypoglycemia exposure. Future studies are required to determine the best dosing periods for chronic SSTR2 antagonism treatment in T2D.

SELECTION OF CITATIONS
SEARCH DETAIL