TY - JOUR A1 - Salvador, Ellaine A1 - Köppl, Theresa A1 - Hörmann, Julia A1 - Schönhärl, Sebastian A1 - Bugaeva, Polina A1 - Kessler, Almuth F. A1 - Burek, Malgorzata A1 - Ernestus, Ralf-Ingo A1 - Löhr, Mario A1 - Hagemann, Carsten T1 - Tumor Treating Fields (TTFields) induce cell junction alterations in a human 3D in vitro model of the blood-brain barrier JF - Pharmaceutics N2 - In a recent study, we showed in an in vitro murine cerebellar microvascular endothelial cell (cerebEND) model as well as in vivo in rats that Tumor-Treating Fields (TTFields) reversibly open the blood–brain barrier (BBB). This process is facilitated by delocalizing tight junction proteins such as claudin-5 from the membrane to the cytoplasm. In investigating the possibility that the same effects could be observed in human-derived cells, a 3D co-culture model of the BBB was established consisting of primary microvascular brain endothelial cells (HBMVEC) and immortalized pericytes, both of human origin. The TTFields at a frequency of 100 kHz administered for 72 h increased the permeability of our human-derived BBB model. The integrity of the BBB had already recovered 48 h post-TTFields, which is earlier than that observed in cerebEND. The data presented herein validate the previously observed effects of TTFields in murine models. Moreover, due to the fact that human cell-based in vitro models more closely resemble patient-derived entities, our findings are highly relevant for pre-clinical studies. KW - blood-brain barrier KW - Tumor-Treating Fields (TTFields) KW - CNS disorders KW - human brain microvascular endothelial cells (HBMVEC) KW - human cells KW - 3D in vitro model Y1 - 2023 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-304830 SN - 1999-4923 VL - 15 IS - 1 ER - TY - THES A1 - Sun, Aili T1 - Effect of Tjap1 knock-down on blood-brain barrier properties under normal and hypoxic conditions T1 - Auswirkung des Tjap1-Knockdowns auf die Eigenschaften der Blut-Hirn-Schranke unter normalen und hypoxischen Bedingungen N2 - Stroke is one of the leading causes of mortality and disability worldwide. The blood-brain barrier (BBB) plays an important role in maintaining brain homeostasis by tightly regulating the exchange of substances between circulating blood and brain parenchyma. BBB disruption is a common pathologic feature of stroke and traumatic brain injury. Understanding the cellular and molecular events that affect the BBB after ischaemic brain injury is important to improve patient prognosis. We have previously shown that microRNA-212/132 is elevated in hypoxic brain microvascular endothelial cells and acts through suppressing the expression of direct microRNA-212/132 target genes with function at the BBB: claudin-1, junctional adhesion molecule 3 (Jam3) and tight-junction associated protein 1 (Tjap1). While the role of claudin-1 and Jam3 at the BBB is well known, the role of Tjap1 is still unclear. The aim of this work was therefore to characterize the role of Tjap1 in brain endothelial cells using a knock-down (KD) approach in established murine in vitro BBB models cEND and cerebEND. Tjap1 KD was established by stable transfection of a plasmid expressing shRNA against Tjap1. The successful downregulation of Tjap1 mRNA and protein was demonstrated by qPCR and Western blot. Tjap1 KD resulted in impaired barrier properties of endothelial cells as shown by lower TEER values and higher paracellular permeability. Interestingly, the Tjap1 KD cells showed lower cell viability and proliferation but migrated faster in a wound healing assay. In the tube formation assay, Tjap1 KD cell lines showed a lower angiogenic potential due to a significantly lower tube length and number as well as a lower amount of branching points in formed capillaries. Tjap1 KD cells showed changes in gene and protein expression. The TJ proteins claudin-5, Jam3 and ZO-1 were significantly increased in Tjap1 KD cell lines, while occludin was strongly decreased. In addition, efflux pump P-glycoprotein was downregulated in Tjap1 KD cells. Oxygen-glucose deprivation (OGD) is a method to mimic stroke in vitro. Brain endothelial cell lines treated with OGD showed lower barrier properties compared to cells cultured under normal condition. These effects were more severe in Tjap1 KD cells, indicating active Tjap1 involvement in the OGD response in brain microvascular endothelial cells. We thus have shown that Tjap1 contributes to a tight barrier of the BBB, regulates cell viability and proliferation of endothelial cells, suppresses their migration and promotes new vessel formation. This means that Tjap1 function is important for mature BBB structure in health and disease. N2 - Schlaganfall ist weltweit eine der häufigsten Ursachen für Mortalität und Behinderung. Die Blut-Hirn-Schranke (BHS) spielt eine wichtige Rolle bei der Aufrechterhaltung der Gehirnhomöostase, indem sie den Stoffaustausch zwischen dem zirkulierenden Blut und dem Gehirnparenchym streng reguliert. Eine Störung der BHS ist ein gemeinsames pathologisches Merkmal von Schlaganfällen und traumatischen Hirnverletzungen. Um die Prognose der Patientinnen und Patienten zu verbessern, ist es wichtig, die zellulären und molekularen Ereignisse zu verstehen, die sich nach einer ischämischen Hirnverletzung auf die BHS auswirken. Wir haben zuvor gezeigt, dass microRNA-212/132 in hypoxischen mikrovaskulären Endothelzellen erhöht ist und durch die Unterdrückung der Expression direkter Zielgene mit Funktion and der BHS wirkt. Zu den Zielgenen von microRNA-212/132 gehören: Claudin-1, Junctional Adhesion Molecule 3 (Jam3) und Tight Junction Associated Protein 1 (Tjap1). Während die Rolle von Caludin-1 und Jam3 and der BHS gut bekannt ist, ist die Rolle von Tjap1 noch unklar. Ziel dieser Arbeit war es daher, die Rolle von Tjap1 in Endothelzellen mithilfe eines Knock-down (KD)-Ansatzes in etablierten murinen In-vitro-BHS-Modellen zu charakterisieren. Tjap1-KD wurde durch stabile Transfektion eines Plasmids etabliert, das shRNA gegen Tjap1 exprimiert. Die erfolgreiche Herunterregulierung von Tjap1-mRNA und -Protein wurde durch qPCR und Western Blot nachgewiesen. Tjap1-KD führte zu einer Beeinträchtigung der Barriereeigenschaften von Endothelzellen, was sich in niedrigeren TEER-Werten und einer höheren parazellulären Permeabilität wiederspiegelte. Interessanterweise zeigten die Tjap1-KD-Zellen in einem Wundheilungstest eine geringere Zelllebensfähigkeit und Proliferation, wanderten jedoch schneller. Im tube formation assay zeigten Tjap1-KD-Zelllinien ein geringeres Angiogenese-Potential durch eine signifikant geringere Anzahl der gebildeten Kapillaren. Tjap-1-KD-Zellen zeigten Veränderungen in der Gen- und Proteinexpression. Die TJ-Proteinen Claudin-5, Jam3 und ZO-1 waren in Tjap1-KD-Zelllinien signifikant erhöht, während Occludin stark verringert war. Darüber hinaus wurde P-Glykoprotein in Tjap1-KD-Zellen herunterreguliert. Sauerstoff-Glukose-Entzug (eng. oxygen/glucose-deprivation, OGD) ist eine Methode zur Nachahmung eines Schlaganfall in vitro. Mit OGD behandelte Endothelzelllinien zeigten im Vergleich zu unter normalen Bedingungen kultivierten Zellen geringere Barriereeigenschaften. Diese Effekte waren in Tjap1-KD-Zellen schwerwiegender, was auf eine aktive Beteiligung von Tjap1 an der OGD-Antwort in Endothelzellen hinweist. Wir haben gezeigt, dass Tjap1 zu einer dichten Barriere der BHS beiträgt, die Zellviabilität und die Proliferation von Endothelzellen reguliert, deren Migration unterdrückt und die Bildung neuer Gefäße fördert. Dies bedeutet, dass die Tjap1-Funktion für die reife BHS-Struktur unter physiologischen und pathophysiologischen Bedingungen wichtig ist. KW - Schlaganfall KW - Blut-Hirn-Schranke KW - blood-brain barrier KW - Tjap1 KW - oxygen/glucose deprivation KW - stroke Y1 - 2023 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-346450 ER - TY - JOUR A1 - Curtaz, Carolin J. A1 - Reifschläger, Leonie A1 - Strähle, Linus A1 - Feldheim, Jonas A1 - Feldheim, Julia J. A1 - Schmitt, Constanze A1 - Kiesel, Matthias A1 - Herbert, Saskia-Laureen A1 - Wöckel, Achim A1 - Meybohm, Patrick A1 - Burek, Malgorzata T1 - Analysis of microRNAs in exosomes of breast cancer patients in search of molecular prognostic factors in brain metastases JF - International Journal of Molecular Sciences N2 - Brain metastases are the most severe tumorous spread during breast cancer disease. They are associated with a limited quality of life and a very poor overall survival. A subtype of extracellular vesicles, exosomes, are sequestered by all kinds of cells, including tumor cells, and play a role in cell-cell communication. Exosomes contain, among others, microRNAs (miRs). Exosomes can be taken up by other cells in the body, and their active molecules can affect the cellular process in target cells. Tumor-secreted exosomes can affect the integrity of the blood-brain barrier (BBB) and have an impact on brain metastases forming. Serum samples from healthy donors, breast cancer patients with primary tumors, or with brain, bone, or visceral metastases were used to isolate exosomes and exosomal miRs. Exosomes expressed exosomal markers CD63 and CD9, and their amount did not vary significantly between groups, as shown by Western blot and ELISA. The selected 48 miRs were detected using real-time PCR. Area under the receiver-operating characteristic curve (AUC) was used to evaluate the diagnostic accuracy. We identified two miRs with the potential to serve as prognostic markers for brain metastases. Hsa-miR-576-3p was significantly upregulated, and hsa-miR-130a-3p was significantly downregulated in exosomes from breast cancer patients with cerebral metastases with AUC: 0.705 and 0.699, respectively. Furthermore, correlation of miR levels with tumor markers revealed that hsa-miR-340-5p levels were significantly correlated with the percentage of Ki67-positive tumor cells, while hsa-miR-342-3p levels were inversely correlated with tumor staging. Analysis of the expression levels of miRs in serum exosomes from breast cancer patients has the potential to identify new, non-invasive, blood-borne prognostic molecular markers to predict the potential for brain metastasis in breast cancer. Additional functional analyzes and careful validation of the identified markers are required before their potential future diagnostic use. KW - breast cancer KW - breast cancer metastases KW - blood-brain barrier KW - patient serum KW - exosomes KW - microRNA KW - gene expression KW - prognostic marker Y1 - 2022 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-284476 SN - 1422-0067 VL - 23 IS - 7 ER - TY - JOUR A1 - Curtaz, Carolin J. A1 - Kiesel, Ludwig A1 - Meybohm, Patrick A1 - Wöckel, Achim A1 - Burek, Malgorzata T1 - Anti-hormonal therapy in breast cancer and its effect on the blood-brain barrier JF - Cancers N2 - Simple Summary Anti-hormonal therapie regimes are well established in oncological treatments in breast cancer. In contrast there is limited knowledge of their effects on metastatic brain metastases in advanced breast cancer and their ability to cross the blood brain-barrier. In this review, we point out the usual antihormonal therapy options in the primary disease, but also in metastatic breast cancer. In addition, we explain the epidemiological facts of brain metastases, as well as the basics of the blood-brain barrier and how this is overcome by metastase. Last but not least, we deal with the known anti-hormonal therapy options and present clinical studies on their intracerebral effect, as well as the known basics of their blood-brain barrier penetration. Not all common anti-hormonal therapeutics are able to penetrate the CNS. It is therefore important for the treating oncologists to use substances that have been proven to cross the BBB, despite the limited data available. Aromataseinhibitors, especially letrozole, probably also tamoxifen, everolimus and CDK4/6 inhibitors, especially abemaciclib, appear to act intracerebrally by overcoming the blood-brain barrier. Nevertheless, further data must be obtained in basic research, but also health care research in relation to patients with brain metastases. Abstract The molecular receptor status of breast cancer has implications for prognosis and long-term metastasis. Although metastatic luminal B-like, hormone-receptor-positive, HER2−negative, breast cancer causes brain metastases less frequently than other subtypes, though tumor metastases in the brain are increasingly being detected of this patient group. Despite the many years of tried and tested use of a wide variety of anti-hormonal therapeutic agents, there is insufficient data on their intracerebral effectiveness and their ability to cross the blood-brain barrier. In this review, we therefore summarize the current state of knowledge on anti-hormonal therapy and its intracerebral impact and effects on the blood-brain barrier in breast cancer. KW - anti-hormonal therapy KW - brain-metastasis KW - blood-brain barrier KW - breast cancer Y1 - 2022 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-290320 SN - 2072-6694 VL - 14 IS - 20 ER - TY - JOUR A1 - Sun, Aili A1 - Blecharz-Lang, Kinga G. A1 - Małecki, Andrzej A1 - Meybohm, Patrick A1 - Nowacka-Chmielewska, Marta M. A1 - Burek, Malgorzata T1 - Role of microRNAs in the regulation of blood-brain barrier function in ischemic stroke and under hypoxic conditions in vitro JF - Frontiers in Drug Delivery N2 - The blood-brain barrier (BBB) is a highly specialized structure that separates the brain from the blood and allows the exchange of molecules between these two compartments through selective channels. The breakdown of the BBB is implicated in the development of severe neurological diseases, especially stroke and traumatic brain injury. Oxygen-glucose deprivation is used to mimic stroke and traumatic brain injury in vitro. Pathways that trigger BBB dysfunction include an imbalance of oxidative stress, excitotoxicity, iron metabolism, cytokine release, cell injury, and cell death. MicroRNAs are small non-coding RNA molecules that regulate gene expression and are emerging as biomarkers for the diagnosis of central nervous system (CNS) injuries. In this review, the regulatory role of potential microRNA biomarkers and related therapeutic targets on the BBB is discussed. A thorough understanding of the potential role of various cellular and linker proteins, among others, in the BBB will open further therapeutic options for the treatment of neurological diseases. KW - blood-brain barrier KW - microRNA KW - stroke KW - traumatic brain injury KW - tight junctions KW - transporter Y1 - 2022 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-291423 SN - 2674-0850 VL - 2 ER - TY - JOUR A1 - Gabbert, Lydia A1 - Dilling, Christina A1 - Meybohm, Patrick A1 - Burek, Malgorzata T1 - Deletion of Protocadherin Gamma C3 Induces Phenotypic and Functional Changes in Brain Microvascular Endothelial Cells In Vitro JF - Frontiers in Pharmacology N2 - Inflammation of the central nervous system (CNS) is associated with diseases such as multiple sclerosis, stroke and neurodegenerative diseases. Compromised integrity of the blood-brain barrier (BBB) and increased migration of immune cells into the CNS are the main characteristics of brain inflammation. Clustered protocadherins (Pcdhs) belong to a large family of cadherin-related molecules. Pcdhs are highly expressed in the CNS in neurons, astrocytes, pericytes and epithelial cells of the choroid plexus and, as we have recently demonstrated, in brain microvascular endothelial cells (BMECs). Knockout of a member of the Pcdh subfamily, PcdhgC3, resulted in significant changes in the barrier integrity of BMECs. Here we characterized the endothelial PcdhgC3 knockout (KO) cells using paracellular permeability measurements, proliferation assay, wound healing assay, inhibition of signaling pathways, oxygen/glucose deprivation (OGD) and a pro-inflammatory cytokine tumor necrosis factor alpha (TNFα) treatment. PcdhgC3 KO showed an increased paracellular permeability, a faster proliferation rate, an altered expression of efflux pumps, transporters, cellular receptors, signaling and inflammatory molecules. Serum starvation led to significantly higher phosphorylation of extracellular signal-regulated kinases (Erk) in KO cells, while no changes in phosphorylated Akt kinase levels were found. PcdhgC3 KO cells migrated faster in the wound healing assay and this migration was significantly inhibited by respective inhibitors of the MAPK-, β-catenin/Wnt-, mTOR- signaling pathways (SL327, XAV939, or Torin 2). PcdhgC3 KO cells responded stronger to OGD and TNFα by significantly higher induction of interleukin 6 mRNA than wild type cells. These results suggest that PcdhgC3 is involved in the regulation of major signaling pathways and the inflammatory response of BMECs. KW - blood-brain barrier KW - protocadherin gamma C3 KW - inflammation KW - oxygen/glucose deprivation KW - stroke KW - tumor necrosis factor-α KW - proliferation Y1 - 2020 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-219828 SN - 1663-9812 VL - 11 ER - TY - JOUR A1 - Schuhmann, Michael K. A1 - Stoll, Guido A1 - Papp, Lena A1 - Bohr, Arne A1 - Volkmann, Jens A1 - Fluri, Felix T1 - Electrical stimulation of the mesencephalic locomotor region has no impact on blood–brain barrier alterations after cerebral photothrombosis in rats JF - International Journal of Molecular Science N2 - Blood–brain barrier (BBB) disruption is a critical event after ischemic stroke, which results in edema formation and hemorrhagic transformation of infarcted tissue. BBB dysfunction following stroke is partly mediated by proinflammatory agents. We recently have shown that high frequency stimulation of the mesencephalic locomotor region (MLR-HFS) exerts an antiapoptotic and anti-inflammatory effect in the border zone of cerebral photothrombotic stroke in rats. Whether MLR-HFS also has an impact on BBB dysfunction in the early stage of stroke is unknown. In this study, rats were subjected to photothrombotic stroke of the sensorimotor cortex and implantation of a stimulating microelectrode into the ipsilesional MLR. Thereafter, either HFS or sham stimulation of the MLR was applied for 24 h. After scarifying the rats, BBB disruption was assessed by determining albumin extravasation and tight junction integrity (claudin 3, claudin 5, and occludin) using Western blot analyses and immunohistochemistry. In addition, by applying zymography, expression of pro-metalloproteinase-9 (pro-MMP-9) was analyzed. No differences were found regarding infarct size and BBB dysfunction between stimulated and unstimulated animals 24 h after induction of stroke. Our results indicate that MLR-HFS neither improves nor worsens the damaged BBB after stroke. Attenuating cytokines/chemokines in the perilesional area, as mediated by MLR-HFS, tend to play a less significant role in preventing the BBB integrity. KW - photothrombotic stroke KW - deep brain stimulation KW - mesencephalic locomotor region KW - blood-brain barrier KW - tight junctions Y1 - 2019 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-201284 SN - 1422-0067 VL - 20 IS - 16 ER - TY - JOUR A1 - Gomes, Sara F. Martins A1 - Westermann, Alexander J. A1 - Sauerwein, Till A1 - Hertlein, Tobias A1 - Förstner, Konrad U. A1 - Ohlsen, Knut A1 - Metzger, Marco A1 - Shusta, Eric V. A1 - Kim, Brandon J. A1 - Appelt-Menzel, Antje A1 - Schubert-Unkmeir, Alexandra T1 - Induced pluripotent stem cell-derived brain endothelial cells as a cellular model to study Neisseria meningitidis infection JF - Frontiers in Microbiology N2 - Meningococcal meningitis is a severe central nervous system infection that occurs when Neisseria meningitidis (Nm) penetrates brain endothelial cells (BECs) of the meningeal blood-cerebrospinal fluid barrier. As a human-specific pathogen, in vivo models are greatly limited and pose a significant challenge. In vitro cell models have been developed, however, most lack critical BEC phenotypes limiting their usefulness. Human BECs generated from induced pluripotent stem cells (iPSCs) retain BEC properties and offer the prospect of modeling the human-specific Nm interaction with BECs. Here, we exploit iPSC-BECs as a novel cellular model to study Nm host-pathogen interactions, and provide an overview of host responses to Nm infection. Using iPSC-BECs, we first confirmed that multiple Nm strains and mutants follow similar phenotypes to previously described models. The recruitment of the recently published pilus adhesin receptor CD147 underneath meningococcal microcolonies could be verified in iPSC-BECs. Nm was also observed to significantly increase the expression of pro-inflammatory and neutrophil-specific chemokines IL6, CXCL1, CXCL2, CXCL8, and CCL20, and the secretion of IFN-γ and RANTES. For the first time, we directly observe that Nm disrupts the three tight junction proteins ZO-1, Occludin, and Claudin-5, which become frayed and/or discontinuous in BECs upon Nm challenge. In accordance with tight junction loss, a sharp loss in trans-endothelial electrical resistance, and an increase in sodium fluorescein permeability and in bacterial transmigration, was observed. Finally, we established RNA-Seq of sorted, infected iPSC-BECs, providing expression data of Nm-responsive host genes. Altogether, this model provides novel insights into Nm pathogenesis, including an impact of Nm on barrier properties and tight junction complexes, and suggests that the paracellular route may contribute to Nm traversal of BECs. KW - Neisseria meningitidis KW - meningococcus KW - bacteria KW - stem cells KW - blood-cerebrospinal fluid barrier KW - blood-brain barrier KW - brain endothelial cells Y1 - 2019 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-201562 VL - 10 IS - 1181 ER - TY - JOUR A1 - Schuhmann, Michael K. A1 - Fluri, Felix T1 - Effects of fullerenols on mouse brain microvascular endothelial cells JF - International Journal of Molecular Sciences N2 - Fullerenols, water-soluble C60-fullerene derivatives, have been shown to exert neuroprotective effects in vitro and in vivo, most likely due to their capability to scavenge free radicals. However, little is known about the effects of fullerenols on the blood–brain barrier (BBB), especially on cerebral endothelial cells under inflammatory conditions. Here, we investigated whether the treatment of primary mouse brain microvascular endothelial cells with fullerenols impacts basal and inflammatory blood–brain barrier (BBB) properties in vitro. While fullerenols (1, 10, and 100 µg/mL) did not change transendothelial electrical resistance under basal and inflammatory conditions, 100 µg/mL of fullerenol significantly reduced erk1/2 activation and resulted in an activation of NFκB in an inflammatory milieu. Our findings suggest that fullerenols might counteract oxidative stress via the erk1/2 and NFκB pathways, and thus are able to protect microvascular endothelial cells under inflammatory conditions. KW - mouse brain microvascular endothelial cell cultur KW - adhesion molecules KW - fullerenes KW - blood-brain barrier KW - inflammation KW - tight junctions Y1 - 2017 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-158072 SN - 1422-0067 VL - 18 IS - 8 ER - TY - JOUR A1 - Shityakov, Sergey A1 - Salvador, Ellaine A1 - Pastorin, Giorgia A1 - Förster, Carola T1 - Blood-brain barrier transport studies, aggregation, and molecular dynamics simulation of multiwalled carbon nanotube functionalized with fluorescein isothiocyanate JF - International Journal of Nanomedicine N2 - In this study, the ability of a multiwalled carbon nanotube functionalized with fluorescein isothiocyanate (MWCNT-FITC) was assessed as a prospective central nervous system-targeting drug delivery system to permeate the blood-brain barrier. The results indicated that the MWCNT-FITC conjugate is able to penetrate microvascular cerebral endothelial monolayers; its concentrations in the Transwell® system were fully equilibrated after 48 hours. Cell viability test, together with phase-contrast and fluorescence microscopies, did not detect any signs of MWCNT-FITC toxicity on the cerebral endothelial cells. These microscopic techniques also revealed presumably the intracellular localization of fluorescent MWCNT-FITCs apart from their massive nonfluorescent accumulation on the cellular surface due to nanotube lipophilic properties. In addition, the 1,000 ps molecular dynamics simulation in vacuo discovered the phenomenon of carbon nanotube aggregation driven by van der Waals forces via MWCN-TFITC rapid dissociation as an intermediate phase. KW - endothelial cells KW - cytotoxicity KW - blood-brain barrier KW - fluorescein isothiocyanate KW - aggregation KW - molecular dynamics KW - fluorescence microscopy KW - Transwell® system KW - multiwalled carbon nanotube KW - mice Y1 - 2015 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-149233 VL - 10 ER -