Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Annika K. Andersson is active.

Publication


Featured researches published by Annika K. Andersson.


Journal of Pineal Research | 2001

Melatonin protects against streptozotocin, but not Interleukin-1β-induced damage of rodent pancreatic β-cells

Annika K. Andersson; Stellan Sandler

In the present study, we examined whether melatonin can protect rodent pancreatic islets against streptozotocin (STZ) and interleukin‐1β (IL‐1β)‐induced suppression of β‐cell function. Formation of free radicals, DNA damage and extensive DNA repair leading to depletion of intracellular nicotinamide adenine dinucleotide (NAD) may mediate STZ toxicity. Activation of inducible nitric oxide synthase and nitric oxide (NO) formation may cause IL‐1β‐induced β‐cell impairment. We also studied the effect of melatonin against STZ‐induced hyperglycemia in C57BL/Ks mice. For in vitro studies, cultured rat islets were exposed to melatonin (100 μM–1 mM) 30 min prior to STZ (0.5 mM) or IL‐1β (25 U/mL) addition. After an additional 30 min incubation with STZ, islet function and NAD content were analyzed either acutely or after 18 hr of recovery in fresh culture medium. For IL‐1β experiments, islets were incubated for 48 hr with the cytokine before evaluation of islet function. We found that melatonin counteracted STZ‐induced inhibition of glucose metabolism and insulin release in cultured rat islets after 18 hr of recovery. Moreover, NAD levels were higher in the melatonin‐treated group at this time point. Melatonin had no effect on IL‐1β‐induced islet inhibition of glucose oxidation or NO formation. Diabetes induced by STZ (140 mg/kg body weight; i.v.) was effectively prevented by administration of melatonin (100 mg/kg body weight; i.p.) 30 min before STZ injection. We conclude that the protective effects of melatonin against β‐cell damage may be related to interference with DNA damage and poly(ADP‐ribose) polymerase (PARP) activation rather than through effects on NO generation pathways.


Upsala Journal of Medical Sciences | 2000

Novel experimental strategies to prevent the development of type 1 diabetes mellitus.

Stellan Sandler; Annika K. Andersson; Andreea Barbu; Claes Hellerström; Maria Holstad; Ella Karlsson; Jan-Olov Sandberg; Eva Strandell; Johan Saldeen; Johnny Sternesjö; Linda Tillmar; Decio L. Eizirik; Malin Flodström; Nils Welsh

Abstract Type 1 diabetes is an autoimmune disease leading to extensive destruction of the pancreatic β-cells. Our research focusses on the role of β-cells during the course of the disease, aiming at finding novel strategies to enhance β-cell resistance against the cytotoxic damage inflicted by the immune system. Special attention has been paid to the possibility that cytokines released by the immune cells infiltrating the pancreatic islets can directly suppress and kill β-cells. Certain cytokines (interleukin-1β, tumor necrosis factor-α and interferon-γ) either alone or in combination, are able to activate signal transduction pathways in β-cells leading to transcription factor activation and de novo gene expression. In this context, it has been found that induction of inducible nitric oxide synthase mediates an elevated production of nitric oxide, which impairs mitochondrial function and causes DNA damage eventually leading to apoptosis and necrosis. However, other induced proteins SUCH AS heat shock protein 70 and superoxide dismutase may reflect a defense reaction elicited in the β-cells by the cytokines. Our strategy is to further seek for proteins involved in both destruction and protection of β-cells. Based on this knowledge, we plan to apply gene therapeutic approaches to increase expression of protective genes in β-cells. If this is feasible we will then evaluate the function and survival of such modified β-cells in animal models of type 1 diabetes such as the NOD mouse. The long-term goal for this research line is to find novel approaches to influence β-cell resistance in humans at risk of developing type 1 diabetes.


Molecular and Cellular Endocrinology | 2005

Cytokines affect PDX-1 expression, insulin and proinsulin secretion from iNOS deficient murine islets.

Annika K. Andersson; Andreas Börjesson; Johanna Sandgren; Stellan Sandler

In rodent islets, exposure to interleukin-1beta (IL-1beta) and interferon-gamma (IFN-gamma) induces expression of inducible nitric oxide synthase (iNOS) and subsequent nitric oxide (NO) formation, which may inhibit islet function. However, cytokines may also induce NO-independent islet suppression. The present aim was to investigate the effect of cytokine exposure to iNOS deficient (iNOS-/-) mouse islets on various islet functions. Islets from iNOS-/- and wt mice exposed to IL-1beta or (IL-1beta + IFN-gamma) for 2-20 h showed different kinetics of glucose-stimulated insulin secretion. In iNOS-/- islets, IL-1beta at high glucose induced a delayed and prolonged stimulation of insulin secretion, and this was followed by an increase in phospholipase D mRNA expression. After 6 and 24 h, proinsulin convertase 1 and 2 (PC1 and PC2) mRNA expression was suppressed and proinsulin secretion increased from wt islets. In iNOS-/- islets, PC1 expression was recovered after 24 h, and there was no difference in proinsulin secretion. PDX-1 mRNA expression was suppressed independent of NO-formation. We conclude that cytokines induce both NO-dependent and NO-independent functional inhibition of murine beta-cells.


Molecular and Cellular Endocrinology | 2004

Cytokine-induced PGE2 formation is reduced from iNOS deficient murine islets

Annika K. Andersson; Lina Thorvaldson; Carina Carlsson; Stellan Sandler

Cytokines may be involved in islet destruction during Type 1 diabetes. Exposure to interleukin-1beta (IL-1beta) or IL-1beta plus interferon-gamma (IFN-gamma) of rodent islets induces expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2). Subsequent formation of nitric oxide (NO) and prostaglandin E(2) (PGE(2)) may impair beta-cell function. Using iNOS deficient (iNOS -/-) islets, we have further investigated the relation between NO formation and PGE(2) induction. We found that iNOS -/- islets responded with a reduced PGE(2) formation following IL-1beta or (IL-1beta + IFN-gamma) treatment compared to wild-type (wt) islets, while COX-2 mRNA or protein content were unchanged. By the addition of an NO donor together with IL-1beta, PGE(2) formation could be stimulated from iNOS -/- islets. We conclude that the lowered capacity of PGE(2) formation observed from cytokine exposed iNOS -/- islets is due to a decreased stimulation of PGE(2) formation by the COX-2 enzyme in the absence of NO, rather then differences in expressed COX-2 protein.


Cell Transplantation | 2006

Survival of an islet allograft deficient in iNOS after implantation into diabetic NOD mice.

Andreas Börjesson; Annika K. Andersson; Stellan Sandler

Proinflammatory cytokines play a major role in rejection of pancreatic islet allografts and in type 1 diabetes (T1D). In rodent islets, exposure to IL-1β alone or combined with IFN-γ induces expression of inducible nitric oxide synthase (iNOS). Inhibition of iNOS or a deletion of the iNOS gene has been shown to be protective in animal models of T1D. In the present study we tested the hypothesis that transplantation of pancreatic islets deficient in iNOS (iNOS–/–) would permit increased graft survival. Pancreatic islets isolated from wild-type (wt) mice and iNOS–/– mice were allogeneically transplanted beneath the kidney capsule of spontaneously diabetic NOD mice. When blood glucose increased above 12.0 mM after preceding normalization of hyperglycemia, animals were sacrificed. Histological examinations of grafts were performed and graft gene expression was analyzed by real-time PCR. Transplantations of the two types of islets could reverse hyperglycemia and the grafts functioned for on average 1 week posttransplantation. Morphological examination of both types of islet grafts showed immune cell infiltration around and within the grafts. Remaining endocrine cells could be observed in wt and iNOS–/– islet grafts. In the removed grafts iNOS-/islet tissue contained higher mRNA levels of insulin, proinsulin convertases (PC-1 and PC-2), and IL-1β compared to transplanted wt islets. The assessments of insulin, PC-1 and PC-2 mRNAs of the grafts suggest that the iNOS–/– islets may be more resistant to destruction in the transplantation model used; however, this was not sufficient to prolong the period of normoglycemia posttransplantation.


Biochemical Pharmacology | 2008

Possible role of an ischemic preconditioning-like response mechanism in KATP channel opener-mediated protection against streptozotocin-induced suppression of rat pancreatic islet function

Stellan Sandler; Annika K. Andersson; Jenny Larsson; Natalia Makeeva; Therese Olsen; Per Arkhammar; John Bondo Hansen; F. Anders Karlsson; Nils Welsh

Potassium channel openers (KCOs) decrease insulin secretion from beta-cells. Some KCOs also protect against damage to beta-cell function and type 1 diabetes in animal models. Previously we have found that the KCO NNC 55-0118 counteracted islet cell dysfunction, and this was associated with a lowering of the mitochondrial membrane potential (Deltapsi). Presently we aimed to explore whether inhibition of insulin secretion per se or rather inhibition of mitochondrial function correlates to counteraction of beta-cell suppression. For this we used two novel KCOs (NNC 55-0321 and NNC 55-0462), which at certain concentrations have different actions regarding insulin secretion and the Deltapsi, with NNC 55-0321 being a potent inhibitor of Deltapsi and NNC 55-0462 being a potent inhibitor of insulin secretion. At 10 microM NNC 55-0321, but not with NNC 55-0462, the islet ATP content and ATP/ADP ratio was acutely decreased. This was accompanied by a complete protection against streptozotocin-induced suppression of islet insulin secretion using the former KCO. In cardiac research KCOs have been used to induce an ischemic preconditioning (IPC) response. In line with an IPC-like mechanism we found that NNC 55-0321 induced an initial free oxygen radical formation, PKC-epsilon isoform activation and a subsequent phosphorylation of the survival promoting factor Akt. Thus, KCOs may elicit mitochondrial events that resemble classical IPC seen in cardiomyocytes, and this could explain the enhanced islet cell function observed. KCOs with this property may be particularly interesting compounds to study as a rescue therapy during acute episodes of beta-cell suppression/destruction.


Immunology Letters | 2003

Effects of interleukin-15 on suppression of rat pancreatic islets in vitro induced by proinflammatory cytokines

Johan Wallström; Annika K. Andersson; Stellan Sandler

The cytokine IL-15 might contribute to inflammatory processes, but also act as an inhibitor of apoptosis in different cell lines. Furthermore, it has been reported that islet cells express IL-15 after exposure to proinflammatory cytokines, which could indicate a defence reaction. We aimed in this study to investigate if IL-15 could influence cell death and/or functional impairment of rat pancreatic islets induced by in vitro exposure to a combination of cytokines (25 U/ml IL-1beta+1000 U/ml IFN-gamma+1000 U/ml TNF-alpha). The effect of IL-15 itself on the function of rat pancreatic islets was also studied. Isolated rat islets were exposed for 24 h to IL-15 at different concentrations in the presence or absence of the cytokine mixture. The cytokines caused a strong inhibition of glucose-stimulated insulin release and the glucose oxidation rates. IL-15 (0.1-10 ng/ml) could not prevent the functional suppression caused by these effects. The cytokine combination caused a decline in whole islet DNA content and a marked increase in non-viable cells analysed by propidium iodide (PI) and annexin V staining. However, there was no significant decrease in whole islet DNA content when IL-15 (0.1 or 1.0 ng/ml) was present together with the cytokine mixture. On the other hand, IL-15 failed to influence the increase in cell death after PI and annexin V staining. If anything, IL-15 alone had a slight stimulatory effect (glucose oxidation rate) on islet cells. In conclusion, we can not exclude that IL-15 might antagonize some cytokine mediated cell death in islet cells, however, IL-15 fails to counteract functional suppression induced by cytokines.


Biochemical and Biophysical Research Communications | 2001

Cytokine-induced inhibition of insulin release from mouse pancreatic β-cells deficient in inducible nitric oxide synthase

Annika K. Andersson; Malin Flodström; Stellan Sandler


Endocrine Journal | 2005

TRANSPLANTATION OF PANCREATIC ISLETS LACKING INDUCIBLE NITRIC OXIDE SYNTHASE TO DIABETIC NOD MICE

Andreas Börjesson; Annika K. Andersson; Stellan Sandler


Archive | 2003

Role of phospholipase D, insulin and proinsulin convertase gene expression in altered insulin secretion from β-cells deficient in inducible nitric oxide synthase following IL-1β and IFN-γ exposure

Annika K. Andersson; Andreas Börjesson; Stellan Sandler

Collaboration


Dive into the Annika K. Andersson's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge