Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Tech Coloproctol ; 26(8): 655-664, 2022 08.
Article in English | MEDLINE | ID: mdl-35593970

ABSTRACT

BACKGROUND: Pelvic surgery carries an inherent risk of autonomic nerve injury leading to genitourinary and bowel dysfunction due to the close proximity of the superior hypogastric plexus (SHP). The aim of this study was to define the detailed anatomy of SHP and identify its relationship with the vascular landmarks and ureters for pelvic autonomic nerve-preserving surgery. METHODS: A cadaveric study on the detailed anatomy of the SHP was conducted in our surgical anatomy research unit. Between 02/2019 and 10/2019, macroscopic anatomical dissections were performed on 45 fresh adult cadavers (39 male, 6 female). Distances between the SHP, major vascular structures, and other anatomical landmarks were measured. RESULTS: Three types of SHP morphology were observed: mesh (64.8%), single nerve (24.4%), and fiber (10.8%). SHP bifurcation was located inferior to the aortic bifurcation in all cases; however, it was observed cranial to the promontory in 80% of the cases, whereas 18% were caudally and 2% were over the promontory. The closest vessels to the left and right of the SHP bifurcation were the left common iliac vein (LCIV) (86.2%, the mean distance was 8.49 ± 7.97 mm) and the right internal iliac artery (RIIA) (48.2%, mean distance was 13.4 ± 9.79 mm), respectively. At SHP bifurcation level, the lateral edge of the SHP was detected on the LCIV in 22 cases and on the RIIA in 10 cases for the left and right side of the plexus, respectively. The distance between the SHP bifurcation and the ureter was 27.9 mm on the right and 24.2 mm on the left. The width of the left (LHN) and right hypogastric nerves (RHN) were 4.35 mm and 4.62 mm at 2 cm below the SHP bifurcation, respectively. LHN was on the vascular structures in 13 cases, whereas RHN in only 1 case, 2 cm below the SHP bifurcation. CONCLUSIONS: Understanding the location of the SHP, including its relationship with important anatomical landmarks, might prevent iatrogenic injury and reduce postoperative morbidity in the pelvic surgery setting.


Subject(s)
Hypogastric Plexus , Ureter , Adult , Autonomic Pathways , Female , Humans , Iliac Vein , Male , Pelvis/innervation
2.
Diabetologia ; 53(7): 1372-83, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20379810

ABSTRACT

AIMS/HYPOTHESIS: Non-invasive imaging of the pancreatic beta cell mass (BCM) requires the identification of novel and specific beta cell biomarkers. We have developed a systems biology approach to the identification of promising beta cell markers. METHODS: We followed a functional genomics strategy based on massive parallel signal sequencing (MPSS) and microarray data obtained in human islets, purified primary rat beta cells, non-beta cells and INS-1E cells to identify promising beta cell markers. Candidate biomarkers were validated and screened using established human and macaque (Macacus cynomolgus) tissue microarrays. RESULTS: After a series of filtering steps, 12 beta cell-specific membrane proteins were identified. For four of the proteins we selected or produced antibodies targeting specifically the human proteins and their splice variants; all four candidates were confirmed as islet-specific in human pancreas. Two splice variants of FXYD domain containing ion transport regulator 2 (FXYD2), a regulating subunit of the Na(+)-K(+)-ATPase, were identified as preferentially present in human pancreatic islets. The presence of FXYD2gammaa was restricted to pancreatic islets and selectively detected in pancreatic beta cells. Analysis of human fetal pancreas samples showed the presence of FXYD2gammaa at an early stage (15 weeks). Histological examination of pancreatic sections from individuals with type 1 diabetes or sections from pancreases of streptozotocin-treated Macacus cynomolgus monkeys indicated a close correlation between loss of FXYD2gammaa and loss of insulin-positive cells. CONCLUSIONS/INTERPRETATION: We propose human FXYD2gammaa as a novel beta cell-specific biomarker.


Subject(s)
Biomarkers/metabolism , Genomics/methods , Insulin-Secreting Cells/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Blotting, Western , Diabetes Mellitus, Type 1/metabolism , Humans , Immunohistochemistry , In Vitro Techniques , Islets of Langerhans/metabolism , Macaca/metabolism , Tissue Array Analysis
3.
Physiol Genomics ; 39(1): 72-81, 2009 Sep 09.
Article in English | MEDLINE | ID: mdl-19622797

ABSTRACT

Pancreatic islet transplantation as a potential cure for type 1 diabetes (T1D) cannot be scaled up due to a scarcity of human pancreas donors. In vitro expansion of beta-cells from mature human pancreatic islets provides an alternative source of insulin-producing cells. The exact nature of the expanded cells produced by diverse expansion protocols and their potential for differentiation into functional beta-cells remain elusive. We performed a large-scale meta-analysis of gene expression in human pancreatic islet cells, which were processed using three different previously described protocols for expansion and for which redifferentiation was attempted. All three expansion protocols induced dramatic changes in the expression profiles of pancreatic islets; many of these changes are shared among the three protocols. Attempts at redifferentiation of expanded cells induce a limited number of gene expression changes. Nevertheless, these fail to restore a pancreatic islet-like gene expression pattern. Comparison with a collection of public microarray datasets confirmed that expanded cells are highly comparable to mesenchymal stem cells. Genes induced in expanded cells are also enriched for targets of transcription factors important for pluripotency induction. The present data increase our understanding of the active pathways in expanded and redifferentiated islets. Knowledge of the mesenchymal stem cell potential may help development of drug therapeutics to restore beta-cell mass in T1D patients.


Subject(s)
Gene Expression Regulation , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Adult , Cell Proliferation , Embryonic Stem Cells/metabolism , Female , Gene Expression Profiling , Gene Regulatory Networks , Humans , Kinetics , Male , Mesenchymal Stem Cells/metabolism , Middle Aged , Oligonucleotide Array Sequence Analysis , Protein Binding
4.
Diabetologia ; 52(6): 1092-101, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19343319

ABSTRACT

AIMS/HYPOTHESIS: The destruction of pancreatic beta cells leading to type 1 diabetes in humans is thought to occur mainly through apoptosis and necrosis induced by activated macrophages and T cells, and in which secreted cytokines play a significant role. The transcription factor nuclear factor kappa-B (NF-kappaB) plays an important role in mediating the apoptotic action of cytokines in beta cells. We therefore sought to determine the changes in expression of genes modulated by NF-kappaB in human islets exposed to a combination of IL1beta, TNF-alpha and IFN-gamma. METHODS: Microarray and gene set enrichment analysis were performed to investigate the global response of gene expression and pathways modulated in cultured human islets exposed to cytokines. Validation of a panel of NF-kappaB-regulated genes was performed by quantitative RT-PCR. The mechanism of induction of BIRC3 by cytokines was examined by transient transfection of BIRC3 promoter constructs linked to a luciferase gene in MIN6 cells, a mouse beta cell line. RESULTS: Enrichment of several metabolic and signalling pathways was observed in cytokine-treated human islets. In addition to the upregulation of known pro-apoptotic genes, a number of anti-apoptotic genes including BIRC3, BCL2A1, TNFAIP3, CFLAR and TRAF1 were induced by cytokines through NF-kappaB. Significant synergy between the cytokines was observed in NF-kappaB-mediated induction of the promoter of BIRC3 in MIN6 cells. CONCLUSIONS/INTERPRETATION: These findings suggest that, via NF-kappaB activation, cytokines induce a concurrent anti-apoptotic pathway that may be critical for preserving islet integrity and viability during the progression of insulitis in type 1 diabetes.


Subject(s)
Cytokines/pharmacology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , NF-kappa B/metabolism , Animals , Baculoviral IAP Repeat-Containing 3 Protein , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Cell Line , Cells, Cultured , DNA-Binding Proteins/genetics , Humans , Inhibitor of Apoptosis Proteins/genetics , Interferon-gamma/pharmacology , Interleukin-1beta/pharmacology , Mice , Minor Histocompatibility Antigens , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins c-bcl-2/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , TNF Receptor-Associated Factor 1/genetics , Tumor Necrosis Factor-alpha/pharmacology , Ubiquitin-Protein Ligases
5.
Diabetologia ; 48(8): 1510-22, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15991020

ABSTRACT

AIMS/HYPOTHESIS: It is thought that enterovirus infections initiate or facilitate the pathogenetic processes leading to type 1 diabetes. Exposure of cultured human islets to cytolytic enterovirus strains kills beta cells after a protracted period, suggesting a role for secondary virus-induced factors such as cytokines. METHODS: To clarify the molecular mechanisms involved in virus-induced beta cell destruction, we analysed the global pattern of gene expression in human islets. After 48 h, RNA was extracted from three independent human islet preparations infected with coxsackievirus B5 or exposed to interleukin 1beta (50 U/ml) plus interferon gamma (1,000 U/ml), and gene expression profiles were analysed using Affymetrix HG-U133A gene chips, which enable simultaneous analysis of 22,000 probe sets. RESULTS: As many as 13,077 genes were detected in control human islets, and 945 and 1293 single genes were found to be modified by exposure to viral infection and the indicated cytokines, respectively. Four hundred and eighty-four genes were similarly modified by the cytokines and viral infection. CONCLUSIONS/INTERPRETATION: The large number of modified genes observed emphasises the complex responses of human islet cells to agents potentially involved in insulitis. Notably, both cytokines and viral infection significantly (p<0.02) increased the expression of several chemokines, the cytokine IL-15 and the intercellular adhesion molecule ICAM-1, which might contribute to the homing and activation of mononuclear cells in the islets during infection and/or an early autoimmune response. The present results provide novel insights into the molecular mechanisms involved in viral- and cytokine-induced human beta cell dysfunction and death.


Subject(s)
Coxsackievirus Infections/metabolism , Cytokines/pharmacology , Gene Expression Regulation/physiology , Islets of Langerhans/metabolism , Aged , Antigen Presentation/genetics , Autoantigens/immunology , Cell Death/genetics , Cell Survival/drug effects , Coxsackievirus Infections/genetics , DNA Repair/genetics , Female , Gene Expression Regulation/drug effects , Humans , Immunohistochemistry , In Vitro Techniques , Inflammation/genetics , Islets of Langerhans/drug effects , Male , Membrane Glycoproteins/genetics , Middle Aged , Multigene Family , Nitrites/metabolism , Oligonucleotide Array Sequence Analysis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Cell Surface/genetics , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptors
6.
Diabetologia ; 46(12): 1641-57, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14600816

ABSTRACT

AIMS/HYPOTHESIS: Viral infections and local production of IFN-gamma might contribute to beta-cell dysfunction/death in Type 1 Diabetes. Double stranded RNA (dsRNA) accumulates in the cytosol of viral-infected cells, and exposure of purified rat beta cells to dsRNA (tested in the form of polyinosinic-polycytidylic acid, PIC) in combination with IFN-gamma results in beta-cell dysfunction and apoptosis. To elucidate the molecular mechanisms involved in PIC + IFN-gamma-effects, we determined the global profile of genes modified by these agents in primary rat beta cells. METHODS: FACS-purified rat beta cells were cultured for 6 or 24 h in control condition or with IFN-gamma, PIC or a combination of both agents. The gene expression profile was analysed in duplicate by high-density oligonucleotide arrays representing 5000 full-length genes and 3000 EST's. Changes of greater than or equal to 2.5-fold were considered as relevant. RESULTS: Following a 6- or 24-h treatment with IFN-gamma, PIC or IFN-gamma and PIC, we observed changes in the expression of 51 to 189 genes. IFN-gamma modified the expression of MHC-related genes, and also of genes involved in beta-cell metabolism, protein processing, cytokines and signal transduction. PIC affected preferentially the expression of genes related to cell adhesion, cytokines and dsRNA signal transduction, transcription factors and MHC. PIC and/or IFN-gamma up-regulated the expression of several chemokines and cytokines that could contribute to mononuclear cell homing and activation during viral infection, while IFN-gamma induced a positive feedback on its own signal transduction. PIC + IFN-gamma inhibited insulin and GLUT-2 expression without modifying pdx-1 mRNA expression. CONCLUSION/INTERPRETATION: This study provides the first comprehensive characterization of the molecular responses of primary beta cells to dsRNA + IFN-gamma, two agents that are probably present in the beta cell milieu during the course of virally-induced insulitis and Type 1 Diabetes. Based on these findings, we propose an integrated model for the molecular mechanisms involved in dsRNA + IFN-gamma induced beta-cell dysfunction and death.


Subject(s)
Gene Expression Profiling , Interferon-gamma/pharmacology , Islets of Langerhans/physiology , RNA, Double-Stranded/pharmacology , Animals , Cells, Cultured , Expressed Sequence Tags , Islets of Langerhans/drug effects , Major Histocompatibility Complex/drug effects , Male , Poly I-C/pharmacology , Rats , Rats, Wistar , Signal Transduction/drug effects , Signal Transduction/physiology
7.
J Biol Chem ; 276(52): 48879-86, 2001 Dec 28.
Article in English | MEDLINE | ID: mdl-11687580

ABSTRACT

Type 1 diabetes mellitus results from an autoimmune destruction of pancreatic beta-cells. Cytokines, such as interleukin-1 beta and interferon-gamma, are putative mediators of immune-induced beta-cell death and, under in vitro conditions, cause beta-cell apoptosis. We have recently shown that interleukin-1 beta + interferon-gamma modifies the expression of >200 genes in beta-cells. Several of these genes are putative targets for the transcription factor nuclear factor-kappa B (NF-kappa B), and in subsequent experiments we showed that NF-kappa B activation is mostly pro-apoptotic in beta-cells. To identify cytokine-induced and NF-kappa B-regulated genes in primary rat beta-cells, we presently combined two experimental approaches: 1) blocking of NF-kappa B activation in cytokine-exposed beta-cells by a recombinant adenovirus (AdI kappa B((SA)2)) containing an inhibitor of NF-kappa B alpha (I kappa Bac) super-repressor (S32A/S36A) and 2) study of gene expression by microarray analysis. We identified 66 cytokine-modified and NF-kappa B-regulated genes in beta-cells. Cytokine-induced NF-kappa B activation decreased Pdx-1 and increased c-Myc expression. This, together with NF-kappa B-dependent inhibition of Glut-2, pro-hormone convertase-1, and Isl-1 expression, probably contributes to the loss of differentiated beta-cell functions. NF-kappa B also regulates several genes encoding for chemokines and cytokines in beta-cells. The present data suggest that NF-kappa B is a key "switch regulator" of transcription factors and gene networks controlling cytokine-induced beta-cell dysfunction and death.


Subject(s)
Cytokines/pharmacology , Gene Expression Regulation , Islets of Langerhans/physiology , NF-kappa B/metabolism , Adenoviridae/genetics , Adenoviridae/metabolism , Animals , Cells, Cultured , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/physiopathology , I-kappa B Proteins/genetics , Islets of Langerhans/drug effects , Male , Models, Biological , NF-kappa B/antagonists & inhibitors , NF-kappa B/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Oligonucleotide Array Sequence Analysis , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL