Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Diabetologia ; 60(6): 1051-1056, 2017 06.
Article in English | MEDLINE | ID: mdl-28299380

ABSTRACT

AIMS/HYPOTHESIS: Endothelial-endocrine cell interactions and vascular endothelial growth factor (VEGF)-A signalling are deemed essential for maternal islet vascularisation, glucose control and beta cell expansion during mouse pregnancy. The aim of this study was to assess whether pregnancy-associated beta cell expansion was affected under conditions of islet hypovascularisation. METHODS: Soluble fms-like tyrosine kinase 1 (sFLT1), a VEGF-A decoy receptor, was conditionally overexpressed in maternal mouse beta cells from 1.5 to 14.5 days post coitum. Islet vascularisation, glycaemic control, beta cell proliferation, individual beta cell size and total beta cell volume were assessed in both pregnant mice and non-pregnant littermates. RESULTS: Conditional overexpression of sFLT1 in beta cells resulted in islet hypovascularisation and glucose intolerance in both pregnant and non-pregnant mice. In contrast to non-pregnant littermates, glucose intolerance in pregnant mice was transient. sFLT1 overexpression did not affect pregnancy-associated changes in beta cell proliferation, individual beta cell size or total beta cell volume. CONCLUSIONS/INTERPRETATION: Reduced intra-islet VEGF-A signalling results in maternal islet hypovascularisation and impaired glycaemic control but does not preclude beta cell expansion during mouse pregnancy.


Subject(s)
Insulin-Secreting Cells/metabolism , Animals , Cell Proliferation/genetics , Cell Proliferation/physiology , Cell Size , Female , Islets of Langerhans/metabolism , Mice , Pregnancy , Rats , Signal Transduction/genetics , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism
2.
Diabetologia ; 59(9): 1834-7, 2016 09.
Article in English | MEDLINE | ID: mdl-27053238

ABSTRACT

The generation of beta(-like) cells to compensate for their absolute or relative shortage in type 1 and type 2 diabetes is an obvious therapeutic strategy. Patients first received grafts of donor islet cells over 25 years ago, but this procedure has not become routine in clinical practice because of a donor cell shortage and (auto)immune problems. Transplantation of differentiated embryonic and induced pluripotent stem cells may overcome some but not all the current limitations. Reprogramming exocrine cells towards functional beta(-like) cells would offer an alternative abundant and autologous source of beta(-like) cells. This review focuses on work by our research group towards achieving such a source of cells. It summarises a presentation given at the 'Can we make a better beta cell?' symposium at the 2015 annual meeting of the EASD. It is accompanied by two other reviews on topics from this symposium (by Amin Ardestani and Kathrin Maedler, DOI: 10.1007/s00125-016-3892-9 , and by Heiko Lickert and colleagues, DOI: 10.1007/s00125-016-3949-9 ) and a commentary by the Session Chair, Shanta Persaud (DOI: 10.1007/s00125-016-3870-2 ).


Subject(s)
Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Pancreas/cytology , Animals , Cell Differentiation/physiology , Humans , Macrophages/metabolism , Transcription Factors/metabolism
3.
J Vis Exp ; (102): e52765, 2015 Aug 07.
Article in English | MEDLINE | ID: mdl-26273954

ABSTRACT

Expansion of pancreatic beta cells in vivo or ex vivo, or generation of beta cells by differentiation from an embryonic or adult stem cell, can provide new expandable sources of beta cells to alleviate the donor scarcity in human islet transplantation as therapy for diabetes. Although recent advances have been made towards this aim, mechanisms that regulate beta cell expansion and differentiation from a stem/progenitor cell remain to be characterized. Here, we describe a protocol for an injury model in the adult mouse pancreas that can function as a tool to study mechanisms of tissue remodeling and beta cell proliferation and differentiation. Partial duct ligation (PDL) is an experimentally induced injury of the rodent pancreas involving surgical ligation of the main pancreatic duct resulting in an obstruction of drainage of exocrine products out of the tail region of the pancreas. The inflicted damage induces acinar atrophy, immune cell infiltration and severe tissue remodeling. We have previously reported the activation of Neurogenin (Ngn) 3 expressing endogenous progenitor-like cells and an increase in beta cell proliferation after PDL. Therefore, PDL provides a basis to study signals involved in beta cell dynamics and the properties of an endocrine progenitor in adult pancreas. Since, it still remains largely unclear, which factors and pathways contribute to beta cell neogenesis and proliferation in PDL, a standardized protocol for PDL will allow for comparison across laboratories.


Subject(s)
Cellular Reprogramming/physiology , Insulin-Secreting Cells/cytology , Pancreas/injuries , Pancreatic Ducts/surgery , Animals , Cell Differentiation/physiology , Cell Proliferation/physiology , Humans , Intraoperative Complications/pathology , Ligation/methods , Male , Mice , Mice, Inbred BALB C , Pancreas/cytology
4.
Stem Cells Transl Med ; 4(6): 555-63, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25848123

ABSTRACT

UNLABELLED: Macrophages are classically considered detrimental for pancreatic ß-cell survival and function, thereby contributing to ß-cell failure in both type 1 (T1D) and 2 (T2D) diabetes mellitus. In addition, adipose tissue macrophages negatively influence peripheral insulin signaling and promote obesity-induced insulin resistance in T2D. In contrast, recent data unexpectedly uncovered that macrophages are not only able to protect ß cells during pancreatitis but also to orchestrate ß-cell proliferation and regeneration after ß-cell injury. Moreover, by altering their activation state, macrophages are able to improve insulin resistance in murine models of T2D. This review will elaborate on current insights in macrophage heterogeneity and on the evolving role of pancreas macrophages during organogenesis, tissue injury, and repair. Additional identification of macrophage subtypes and of their secreted factors might ultimately translate into novel therapeutic strategies for both T1D and T2D. SIGNIFICANCE: Diabetes mellitus is a pandemic disease, characterized by severe acute and chronic complications. Macrophages have long been considered prime suspects in the pathogenesis of both type 1 and 2 diabetes mellitus. In this concise review, current insights in macrophage heterogeneity and on the, as yet, underappreciated role of alternatively activated macrophages in insulin sensing and ß-cell development/repair are reported. Further identification of macrophage subtypes and of their secreted factors might ultimately translate into novel therapeutic strategies for diabetes mellitus.


Subject(s)
Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Macrophages/metabolism , Regeneration , Animals , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/therapy , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/therapy , Humans , Macrophages/pathology , Pancreatitis/metabolism , Pancreatitis/pathology , Pancreatitis/therapy
5.
Eur J Immunol ; 45(5): 1482-93, 2015 May.
Article in English | MEDLINE | ID: mdl-25645754

ABSTRACT

Pancreas injury by partial duct ligation (PDL) activates a healing response, encompassing ß-cell neogenesis and proliferation. Macrophages (MΦs) were recently shown to promote ß-cell proliferation after PDL, but they remain poorly characterized. We assessed myeloid cell diversity and the factors driving myeloid cell dynamics following acute pancreas injury by PDL. In naive and sham-operated pancreas, the myeloid cell compartment consisted mainly of two distinct tissue-resident MΦ types, designated MHC-II(lo) and MHC-II(hi) MΦs, the latter being predominant. MHC-II(lo) and MHC-II(hi) pancreas MΦs differed at the molecular level, with MHC-II(lo) MΦs being more M2-activated. After PDL, there was an early surge of Ly6C(hi) monocyte infiltration in the pancreas, followed by a transient MHC-II(lo) MΦ peak and ultimately a restoration of the MHC-II(hi) MΦ-dominated steady-state equilibrium. These intricate MΦ dynamics in PDL pancreas depended on monocyte recruitment by C-C chemokine receptor 2 and macrophage-colony stimulating factor receptor as well as on macrophage-colony stimulating factor receptor-dependent local MΦ proliferation. Functionally, MHC-II(lo) MΦs were more angiogenic. We further demonstrated that, at least in C-C chemokine receptor 2-KO mice, tissue MΦs, rather than Ly6C(hi) monocyte-derived MΦs, contributed to ß-cell proliferation. Together, our study fully characterizes the MΦ subsets in the pancreas and clarifies the complex dynamics of MΦs after PDL injury.


Subject(s)
Macrophages/immunology , Macrophages/pathology , Monocytes/immunology , Monocytes/pathology , Pancreas/immunology , Pancreas/injuries , Animals , Antigens, Ly/metabolism , Cell Movement/immunology , Cell Proliferation , Cellular Microenvironment/immunology , Histocompatibility Antigens Class II/metabolism , Ligation , Macrophage Activation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Cells/classification , Myeloid Cells/immunology , Myeloid Cells/pathology , Pancreas/pathology , Pancreatic Ducts/injuries , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Regeneration/immunology
7.
Diabetologia ; 57(1): 140-7, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24121626

ABSTRACT

AIMS/HYPOTHESIS: Vascular endothelial growth factor (VEGF) has been recognised by loss-of-function experiments as a pleiotropic factor with importance in embryonic pancreas development and postnatal beta cell function. Chronic, nonconditional overexpression of VEGF-A has a deleterious effect on beta cell development and function. We report, for the first time, a conditional gain-of-function study to evaluate the effect of transient VEGF-A overexpression by adult pancreatic beta cells on islet vasculature and beta cell proliferation and survival, under both normal physiological and injury conditions. METHODS: In a transgenicmouse strain, overexpressing VEGF-A in a doxycycline-inducible and beta cell-specific manner, we evaluated the ability of VEGF-A to affect islet vessel density, beta cell proliferation and protection of the adult beta cell mass from toxin-induced injury. RESULTS: Short-term VEGF-A overexpression resulted in islet hypervascularisation, increased beta cell proliferation and protection from toxin-mediated beta cell death, and thereby prevented the development of hyperglycaemia. Extended overexpression of VEGF-A led to impaired glucose tolerance, elevated fasting glycaemia and a decreased beta cell mass. CONCLUSIONS/INTERPRETATION: Overexpression of VEGF-A in beta cells time-dependently affects glycometabolic control and beta cell protection and proliferation. These data nourish further studies to examine the role of controlled VEGF delivery in (pre)clinical applications aimed at protecting and/or restoring the injured beta cell mass.


Subject(s)
Diabetes Mellitus/prevention & control , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Proliferation , Cell Survival/physiology , Diabetes Mellitus/metabolism , Islets of Langerhans/blood supply , Islets of Langerhans/metabolism , Mice , Mice, Transgenic , Rats , Vascular Endothelial Growth Factor A/genetics
8.
FASEB J ; 25(7): 2433-46, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21478264

ABSTRACT

Accurate determination of tumor human epidermal growth factor receptor 2 (HER2)-status in breast cancer patients is possible via noninvasive imaging, provided adequate tracers are used. In this study, we describe the generation of a panel of 38 nanobodies, small HER2-binding fragments that are derived from heavy-chain-only antibodies raised in an immunized dromedary. In search of a lead compound, a subset of nanobodies was biochemically characterized in depth and preclinically tested for use as tracers for imaging of xenografted tumors. The selected compound, 2Rs15d, was found to be stable and to interact specifically with HER2 recombinant protein and HER2-expressing cells in ELISA, surface plasmon resonance, flow cytometry, and radioligand binding studies with low nanomolar affinities, and did not compete with anti-HER2 therapeutic antibodies trastuzumab and pertuzumab. Single-photon-emission computed tomography (SPECT) imaging quantification and biodistribution analyses showed that (99m)Tc-labeled 2Rs15d has a high tumor uptake in 2 HER2(+) tumor models, fast blood clearance, low accumulation in nontarget organs except kidneys, and high concomitant tumor-to-blood and tumor-to-muscle ratios at 1 h after intravenous injection. These values were dramatically lower for an irrelevant control (99m)Tc-nanobody and for (99m)Tc-2Rs15d targeting a HER2(-) tumor.


Subject(s)
Mammary Neoplasms, Experimental/immunology , Molecular Imaging/methods , Receptor, ErbB-2/immunology , Single-Chain Antibodies/immunology , Animals , Antibody Specificity/immunology , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CHO Cells , Camelids, New World , Cell Line, Tumor , Cricetinae , Cricetulus , Female , Humans , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Nude , Peptide Library , Single-Chain Antibodies/genetics , Single-Chain Antibodies/pharmacokinetics , Technetium/pharmacokinetics , Tissue Distribution , Tomography, Emission-Computed, Single-Photon , Transplantation, Heterologous , X-Ray Microtomography
SELECTION OF CITATIONS
SEARCH DETAIL
...