Network


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

Hotspot


Dive into the research topics where Robert Bränström is active.

Publication


Featured researches published by Robert Bränström.


The EMBO Journal | 1998

Cysteine string protein (CSP) is an insulin secretory granule-associated protein regulating beta-cell exocytosis.

Hilary Brown; Olof Larsson; Robert Bränström; Shao-Nian Yang; Barbara Leibiger; Ingo B. Leibiger; Gabriel Fried; Tilo Moede; Jude T. Deeney; Graham R. Brown; Gunilla Jacobsson; Christopher J. Rhodes; Janice E.A. Braun; Richard H. Scheller; Barbara E. Corkey; Per-Olof Berggren; Björn Meister

Cysteine string proteins (CSPs) are novel synaptic vesicle‐associated protein components characterized by an N‐terminal J‐domain and a central palmitoylated string of cysteine residues. The cellular localization and functional role of CSP was studied in pancreatic endocrine cells. In situ hybridization and RT–PCR analysis demonstrated CSP mRNA expression in insulin‐producing cells. CSP1 mRNA was present in pancreatic islets; both CSP1 and CSP2 mRNAs were seen in insulin‐secreting cell lines. Punctate CSP‐like immunoreactivity (CSP‐LI) was demonstrated in most islets of Langerhans cells, acinar cells and nerve fibers of the rat pancreas. Ultrastructural analysis showed CSP‐LI in close association with membranes of secretory granules of cells in the endo‐ and exocrine pancreas. Subcellular fractionation of insulinoma cells showed CSP1 (34/36 kDa) in granular fractions; the membrane and cytosol fractions contained predominantly CSP2 (27 kDa). The fractions also contained proteins of 72 and 70 kDa, presumably CSP dimers. CSP1 overexpression in INS‐1 cells or intracellular administration of CSP antibodies into mouse ob/ob β‐cells did not affect voltage‐dependent Ca2+‐channel activity. Amperometric measurements showed a significant decrease in insulin exocytosis in individual INS‐1 cells after CSP1 overexpression. We conclude that CSP is associated with insulin secretory granules and that CSP participates in the molecular regulation of insulin exocytosis by mechanisms not involving changes in the activity of voltage‐gated Ca2+‐channels.


Journal of Biological Chemistry | 1998

DIRECT INHIBITION OF THE PANCREATIC BETA -CELL ATP-REGULATED POTASSIUM CHANNEL BY ALPHA -KETOISOCAPROATE

Robert Bränström; Suad Efendic; Per-Olof Berggren; Olof Larsson

The ATP-regulated potassium (K ATP) channel plays an essential role in the control of insulin release from the pancreatic β-cell. In the present study we have used the patch-clamp technique to study the direct effects of α-ketoisocaproate on the K ATPchannel in isolated patches and intact pancreatic β-cells. In excised inside-out patches, the activity of the K ATPchannel was dose-dependently inhibited by α-ketoisocaproate, half-maximal concentration being approximately 8 mm. The blocking effect of α-ketoisocaproate was fully reversible. Stimulation of channel activity by the addition of ATP/ADP (ratio 1) did not counteract the inhibitory effect of α-ketoisocaproate. In the presence of the metabolic inhibitor sodium azide, α-ketoisocaproate was still able to inhibit single channel activity in excised patches and to block whole cell K ATP currents in intact cells. No effect of α-ketoisocaproate could be obtained on either the large or the small conductance Ca2+-regulated K+ channel. Enzymatic treatment of the patches with trypsin prevented the inhibitory effect of α-ketoisocaproate. Based on these observations, it is unlikely that the blocking effect of α-ketoisocaproate is due to an unspecific effect on K+ channel pores. Leucine, the precursor of α-ketoisocaproate, did not affectK ATP channel activity in excised patches. Our findings are compatible with the view that α-ketoisocaproate not only affects the β-cell stimulus secretion coupling by generation of ATP but also by direct inhibition of the K ATPchannel.


Journal of Molecular Endocrinology | 2010

Expression and association of TRPC subtypes with Orai1 and STIM1 in human parathyroid

Ming Lu; Robert Bränström; Erik Berglund; Anders Höög; Peyman Björklund; Gunnar Westin; Catharina Larsson; Lars-Ove Farnebo; Lars Forsberg

The mechanism behind Ca(2)(+) entry into the parathyroid cells has been widely debated, and the molecular identities of the responsible ion channels have not been established yet. In this study, we show that the parathyroid cells lack voltage-operated Ca(2)(+) channels. Passive store depletion by thapsigargin, on the other hand, induces a large non-voltage-activated non-selective cation current. The increase in intracellular Ca(2)(+) caused by thapsigargin is attenuated by 2-aminoethoxydiphenyl borate, a blocker of store-operated Ca(2)(+) entry (SOCE). Candidate molecules for non-voltage-operated Ca(2)(+) signaling were investigated. These included members of the transient receptor potential canonical (TRPC) ion channel family, as well as Ca(2)(+) release-activated Ca(2)(+) modulator 1 (Orai1) and stromal interaction molecule 1 (STIM1) that are key proteins in the SOCE pathway. Using RT-PCR screening, quantitative real-time PCR, and western blot, we showed expression of TRPC1, TRPC4, and TRPC6; Orai1; and STIM1 genes and proteins in normal and adenomatous human parathyroid tissues. Furthermore, co-immunoprecipitation experiments demonstrated a ternary complex of TRPC1-Orai1-STIM1, supporting a physical interaction between these molecules in human parathyroid.


Experimental Cell Research | 2014

Functional role of the Ca2+-activated Cl- channel DOG1/TMEM16A in gastrointestinal stromal tumor cells

Erik Berglund; Pinar Akçakaya; David Berglund; Fredrik Karlsson; Vladana Vukojević; Linkiat Lee; Darko Bogdanović; Weng-Onn Lui; Catharina Larsson; Jan Zedenius; Robin Fröbom; Robert Bränström

DOG1, a Ca(2+)-activated Cl(-) channel (CaCC), was identified in 2004 to be robustly expressed in gastrointestinal stromal tumors (GIST). It was rapidly included as a tumor marker in routine diagnostics, but the functional role remained unknown. CaCCs are important regulators of normal physiological functions, but also implicated in tumorigenesis, cancer progression, metastasis, cell migration, apoptosis, proliferation and viability in several malignancies. We therefore investigated whether DOG1 plays a role in the three latter in GIST by utilizing in vitro cell model systems. Confocal microscopy identified different subcellular localizations of DOG1 in imatinib-sensitive and imatinib-resistant cells. Electrophysiological studies confirmed that DOG1-specific pharmacological agents possess potent activating and inhibiting properties. Proliferation assays showed small effects up to 72 h, and flow cytometric analysis of adherent cells with 7-AAD/Annexin V detected no pharmacological effects on viable GIST cells. However, inhibition of DOG1 conveyed pro-apoptotic effects among early apoptotic imatinib-resistant cells. In conclusion, DOG1 generates Cl(-) currents in GIST that can be regulated pharmacologically, with small effects on cell viability and proliferation in vitro. Inhibition of DOG1 might act pro-apoptotic on some early apoptotic GIST cell populations. Further studies are warranted to fully illuminate the function of DOG1 and its potential as therapeutic target.


American Journal of Physiology-cell Physiology | 2009

BLX-1002, a novel thiazolidinedione with no PPAR affinity, stimulates AMP-activated protein kinase activity, raises cytosolic Ca2+, and enhances glucose-stimulated insulin secretion in a PI3K-dependent manner

Fan Zhang; Deben Dey; Robert Bränström; Lars Forsberg; Ming Lu; Qimin Zhang; Åke Sjöholm

BLX-1002 is a novel small thiazolidinedione with no apparent affinity to peroxisome proliferator-activated receptors (PPAR) that has been shown to reduce glycemia in type 2 diabetes without adipogenic effects. Its precise mechanisms of action, however, remain elusive, and no studies have been done with respect to possible effects of BLX-1002 on pancreatic beta-cells. We have investigated the influence of the drug on beta-cell function in mouse islets in vitro. BLX-1002 enhanced insulin secretion stimulated by high, but not low or intermediate, glucose concentrations. BLX-1002 also augmented cytoplasmic free Ca2+ concentration ([Ca2+](i)) at high glucose, an effect that was abolished by pretreatment with the Ca2+-ATPase inhibitor thapsigargin. In contrast, BLX-1002 did not interfere with voltage-gated Ca2+ channel or ATP-sensitive K+ channel activities. In addition, cellular NAD(P)H stimulated by glucose was not affected by the drug. The stimulatory effect of BLX-1002 on insulin secretion at high glucose was completely abolished by treatment with the phosphatidylinositol 3-kinase (PI3K) inhibitors wortmannin or LY-294002. Stimulation of the beta-cells with BLX-1002 also induced activation of AMP-activated protein kinase (AMPK) at high glucose. Our study suggests that BLX-1002 potentiates insulin secretion only at high glucose in beta-cells in a PI3K-dependent manner. This effect of BLX-1002 is associated with an increased [Ca2+](i) mediated through Ca2+ mobilization, and an enhanced activation of AMPK. The glucose-sensitive stimulatory impact of BLX-1002 on beta-cell function may translate into substantial clinical benefits of the drug in the management of type 2 diabetes, by avoidance of hypoglycemia.


Journal of Intensive Care Medicine | 2015

Pheochromocytoma-Induced Inverted Takotsubo-Like Cardiomyopathy Leading to Cardiogenic Shock Successfully Treated With Extracorporeal Membrane Oxygenation

Benjamin Flam; Michael Broomé; Björn Frenckner; Robert Bränström; Max Bell

Pheochromocytoma classically displays a variety of rather benign symptoms, such as headache, palpitations, and sweating, although severe cardiac manifestations have been described. We report a case of pheochromocytoma-induced inverted takotsubo-like cardiomyopathy leading to shock and cardiac arrest successfully treated with extracorporeal membrane oxygenation (ECMO) as a bridge to pharmacological therapy and curative adrenalectomy. A previously healthy 46-year-old woman presented to the emergency department with abdominal pain, dyspnea, nausea, and vomiting. Clinical evaluation revealed cardiorespiratory failure with hypoxia and severe metabolic acidosis. Computed tomography (CT) scan showed pulmonary edema and a left adrenal mass. Transthoracic echocardiography (TTE) displayed severe left ventricular dysfunction with inverted takotsubo contractile pattern. Despite mechanical ventilation and inotropic and vasopressor support, asystolic cardiac arrest ensued. The patient was resuscitated using manual chest compressions followed by venoarterial ECMO. Repeated TTEs demonstrated resolution of the cardiomyopathy within a few days. Laboratory results indicated transient renal and hepatic dysfunction, and CT scan of the brain displayed occipital infarctions. Biochemical testing and radionuclide scintigraphy confirmed a pheochromocytoma. Pharmacological adrenergic blockade was instituted prior to delayed adrenalectomy after which the diagnosis was histopathologically verified. The patient recovered after rehabilitation. We conclude that pheochromocytoma should be considered in patients presenting with unexplained cardiovascular compromise, especially if they display (inverted) takotsubo contractile pattern. Timely, adequate management might involve ECMO as a bridge to pharmacological therapy and curative surgery.


FEBS Letters | 2007

3H-serotonin as a marker of oscillatory insulin secretion in clonal β-cells (INS-1)

Jude T. Deeney; Robert Bränström; Barbara E. Corkey; Olof Larsson; Per-Olof Berggren

Serotonin release from preloaded pancreatic β‐cells has been used as a marker for insulin release in studying exocytosis from single cells using the amperometric technique. We found that single pancreatic β‐cells exhibited oscillations in exocytosis with a period of 1–1.5 min as measured amperometrically by serotonin release. We also show that 3H‐serotonin can be used to monitor exocytosis from intact and streptolysin‐O permeabilized clonal insulin‐secreting cells preloaded with labeled serotonin and that serotonin release correlated with insulin secretion in the same cells. The use of 3H‐serotonin provides a real‐time indicator of exocytosis from populations of clonal insulin‐secreting cells.


Journal of Biological Chemistry | 2003

High Extracellular Ca2+ Hyperpolarizes Human Parathyroid Cells via Ca2+-activated K+ Channels

Stiina Välimäki; Anders Höög; Catharina Larsson; Lars-Ove Farnebo; Robert Bränström

Membrane potential has a major influence on stimulus-secretion coupling in various excitable cells. The role of membrane potential in the regulation of parathyroid hormone secretion is not known. High K+-induced depolarization increases secretion from parathyroid cells. The paradox is that increased extracellular Ca2+, which inhibits secretion, has also been postulated to have a depolarizing effect. In this study, human parathyroid cells from parathyroid adenomas were used in patch clamp studies of K+ channels and membrane potential. Detailed characterization revealed two K+ channels that were strictly dependent of intracellular Ca2+ concentration. At high extracellular Ca2+, a large K+ current was seen, and the cells were hyperpolarized (–50.4 ± 13.4 mV), whereas lowering of extracellular Ca2+ resulted in a dramatic decrease in K+ current and depolarization of the cells (–0.1 ± 8.8 mV, p < 0.001). Changes in extracellular Ca2+ did not alter K+ currents when intracellular Ca2+ was clamped, indicating that K+ channels are activated by intracellular Ca2+. The results were concordant in cell-attached, perforated patch, whole-cell and excised membrane patch configurations. These results suggest that [Ca2+]o regulates membrane potential of human parathyroid cells via Ca2+-activated K+ channels and that the membrane potential may be of greater importance for the stimulus-secretion coupling than recognized previously.


Anti-Cancer Drugs | 2014

Intracellular concentration of the tyrosine kinase inhibitor imatinib in gastrointestinal stromal tumor cells.

Erik Berglund; Sarojini J. K. A. Ubhayasekera; Fredrik Karlsson; Pinar Akçakaya; Warunika Aluthgedara; Jan Åhlén; Robin Fröbom; Inga-Lena Nilsson; Weng-Onn Lui; Catharina Larsson; Jan Zedenius; Jonas Bergquist; Robert Bränström

Gastrointestinal stromal tumor (GIST) is the most common mesenchymal neoplasm in the gastrointestinal tract. In most GISTs, the underlying mechanism is a gain-of-function mutation in the KIT or the PDGFRA gene. Imatinib is a tyrosine kinase inhibitor that specifically blocks the intracellular ATP-binding sites of these receptors. A correlation exists between plasma levels of imatinib and progression-free survival, but it is not known whether the plasma concentration correlates with the intracellular drug concentration. We determined intracellular imatinib levels in two GIST cell lines: the imatinib-sensitive GIST882 and the imatinib-resistant GIST48. After exposing the GIST cells to imatinib, the intracellular concentrations were evaluated using LC-MS (TOF). The concentration of imatinib in clinical samples from three patients was also determined to assess the validity and reliability of the method in the clinical setting. Determination of imatinib uptake fits within detection levels and values are highly reproducible. The GIST48 cells showed significantly lower imatinib uptake compared with GIST882 in therapeutic doses, indicating a possible difference in uptake mechanisms. Furthermore, imatinib accumulated in the tumor tissues and showed intratumoral regional differences. These data show, for the first time, a feasible and reproducible technique to measure intracellular imatinib levels in experimental and clinical settings. The difference in the intracellular imatinib concentration between the cell lines and clinical samples indicates that drug transporters may contribute toward resistance mechanisms in GIST cells. This highlights the importance of further clinical studies to quantify drug transporter expression and measure intracellular imatinib levels in GIST patients.


Experimental Cell Research | 2013

Evidence for Ca2+-regulated ATP release in gastrointestinal stromal tumors

Erik Berglund; David Berglund; Pinar Akçakaya; Mehran Ghaderi; Elisabetta Daré; Per-Olof Berggren; Martin Köhler; Craig A. Aspinwall; Weng-Onn Lui; Jan Zedenius; Catharina Larsson; Robert Bränström

Gastrointestinal stromal tumors (GISTs) are thought to originate from the electrically active pacemaker cells of the gastrointestinal tract. Despite the presence of synaptic-like vesicles and proteins involved in cell secretion it remains unclear whether GIST cells possess regulated release mechanisms. The GIST tumor cell line GIST882 was used as a model cell system, and stimulus-release coupling was investigated by confocal microscopy of cytoplasmic free Ca(2+) concentration ([Ca(2+)]i), flow cytometry, and luminometric measurements of extracellular ATP. We demonstrate that GIST cells have an intact intracellular Ca(2+)-signaling pathway that regulates ATP release. Cell viability and cell membrane integrity was preserved, excluding ATP leakage due to cell death and suggesting active ATP release. The stimulus-secretion signal transduction is at least partly dependent on Ca(2+) influx since exclusion of extracellular Ca(2+) diminishes the ATP release. We conclude that measurements of ATP release in GISTs may be a useful tool for dissecting the signal transduction pathway, mapping exocytotic components, and possibly for the development and evaluation of drugs. Additionally, release of ATP from GISTs may have importance for tumor tissue homeostasis and immune surveillance escape.

Collaboration


Dive into the Robert Bränström's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Catharina Larsson

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anders Höög

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Pinar Akçakaya

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ming Lu

Karolinska Institutet

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge