Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 9 de 9
1.
J Control Release ; 369: 668-683, 2024 Apr 11.
Article En | MEDLINE | ID: mdl-38548064

Local and long-lasting administration of potent chemotherapeutics is a promising therapeutic intervention to increase the efficiency of chemotherapy of hard-to-treat tumors such as the most lethal brain tumors, glioblastomas (GBM). However, despite high toxicity for GBM cells, potent chemotherapeutics such as gemcitabine (Gem) cannot be widely implemented as they do not efficiently cross the blood brain barrier (BBB). As an alternative method for continuous administration of Gem, we here operate freestanding iontronic pumps - "GemIPs" - equipped with a custom-synthesized ion exchange membrane (IEM) to treat a GBM tumor in an avian embryonic in vivo system. We compare GemIP treatment effects with a topical metronomic treatment and observe that a remarkable growth inhibition was only achieved with steady dosing via GemIPs. Daily topical drug administration (at the maximum dosage that was not lethal for the embryonic host organism) did not decrease tumor sizes, while both treatment regimes caused S-phase cell cycle arrest and apoptosis. We hypothesize that the pharmacodynamic effects generate different intratumoral drug concentration profiles for each technique, which causes this difference in outcome. We created a digital model of the experiment, which proposes a fast decay in the local drug concentration for the topical daily treatment, but a long-lasting high local concentration of Gem close to the tumor area with GemIPs. Continuous chemotherapy with iontronic devices opens new possibilities in cancer treatment: the long-lasting and highly local dosing of clinically available, potent chemotherapeutics to greatly enhance treatment efficiency without systemic side-effects. SIGNIFICANCE STATEMENT: Iontronic pumps (GemIPs) provide continuous and localized administration of the chemotherapeutic gemcitabine (Gem) for treating glioblastoma in vivo. By generating high and constant drug concentrations near the vascularized growing tumor, GemIPs offer an efficient and less harmful alternative to systemic administration. Continuous GemIP dosing resulted in remarkable growth inhibition, superior to daily topical Gem application at higher doses. Our digital modelling shows the advantages of iontronic chemotherapy in overcoming limitations of burst release and transient concentration profiles, and providing precise control over dosing profiles and local distribution. This technology holds promise for future implants, could revolutionize treatment strategies, and offers a new platform for studying the influence of timing and dosing dependencies of already-established drugs in the fight against hard-to-treat tumors.

2.
Proc Natl Acad Sci U S A ; 120(28): e2210152120, 2023 07 11.
Article En | MEDLINE | ID: mdl-37406102

Sepsis has emerged as a global health burden associated with multiple organ dysfunction and 20% mortality rate in patients. Numerous clinical studies over the past two decades have correlated the disease severity and mortality in septic patients with impaired heart rate variability (HRV), as a consequence of impaired chronotropic response of sinoatrial node (SAN) pacemaker activity to vagal/parasympathetic stimulation. However, the molecular mechanism(s) downstream to parasympathetic inputs have not been investigated yet in sepsis, particularly in the SAN. Based on electrocardiography, fluorescence Ca2+ imaging, electrophysiology, and protein assays from organ to subcellular level, we report that impaired muscarinic receptor subtype 2-G protein-activated inwardly-rectifying potassium channel (M2R-GIRK) signaling in a lipopolysaccharide-induced proxy septic mouse model plays a critical role in SAN pacemaking and HRV. The parasympathetic responses to a muscarinic agonist, namely IKACh activation in SAN cells, reduction in Ca2+ mobilization of SAN tissues, lowering of heart rate and increase in HRV, were profoundly attenuated upon lipopolysaccharide-induced sepsis. These functional alterations manifested as a direct consequence of reduced expression of key ion-channel components (GIRK1, GIRK4, and M2R) in the mouse SAN tissues and cells, which was further evident in the human right atrial appendages of septic patients and likely not mediated by the common proinflammatory cytokines elevated in sepsis.


Lipopolysaccharides , Sepsis , Humans , Animals , Mice , Lipopolysaccharides/toxicity , Lipopolysaccharides/metabolism , Sinoatrial Node/physiology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Signal Transduction/physiology , Sepsis/chemically induced , Sepsis/metabolism
3.
Front Oncol ; 12: 998907, 2022.
Article En | MEDLINE | ID: mdl-36483038

A search in the GDC Data Portal revealed 304 documented somatic mutations of the KCNJ3 gene in primary tumors (out of 10.202 cases). Most affected tumor types were carcinomas from uterus, skin and lung, while breast cancer exerted the lowest number of somatic mutations. We focused our research on 15 missense mutations within the region between TM1 and TM2, comprising the pore helix and ion selectivity signature. Expression was measured by confocal laser scan microscopy of eGFP tagged GIRK1 subunits, expressed with and without GIRK4 in oocytes of Xenopus laevis. GIRK ion currents were activated via coexpressed m2Rs and measured by the Two Electrode Voltage Clamp technique. Magnitude of the total GIRK current, as well as the fraction of current inducible by the agonist, were measured. Ion selectivity was gauged by assessment of the PNa+/PK+ ratio, calculated by the GIRK current reversal potential in extracellular media at different Na+ and K+ concentrations. None of the tested mutations was able to form functional GIRK1 homooligomeric ion channels. One of the mutations, G145A, which locates directly to the ion selectivity signature, exerted an increased PNa+/PK+ ratio. Generally, the missense mutations studied can be categorized into three groups: (i) normal/reduced expression accompanied by reduced/absent function (S132Y, F136L, E139K, G145A, R149Q, R149P, G178D, S185Y, Q186R), (ii) normal/increased expression as well as increased function (E140M, A142T, M184I) and (iii) miniscule expression but increased function relative to expression levels (I151N, G158S). We conclude, that gain of function mutations, identical or similar to categories (ii) and (iii), may potentially be involved in genesis and progression of malignancies in tissues that exert a high rate of occurrence of somatic mutations of KCNJ3.

4.
Sci Rep ; 9(1): 19277, 2019 12 17.
Article En | MEDLINE | ID: mdl-31848385

Excessive expression of subunit 1 of GIRK1 in ER+ breast tumors is associated with reduced survival times and increased lymph node metastasis in patients. To investigate possible tumor-initiating properties, benign MCF10A and malign MCF7 mammary epithelial cells were engineered to overexpress GIRK1 neoplasia associated vital parameters and resting potentials were measured and compared to controls. The presence of GIRK1 resulted in resting potentials negative to the controls. Upon GIRK1 overexpression, several cellular pathways were regulated towards pro-tumorigenic action as revealed by comparison of transcriptomes of MCF10AGIRK1 with the control (MCF10AeGFP). According to transcriptome analysis, cellular migration was promoted while wound healing and extracellular matrix interactions were impaired. Vital parameters in MCF7 cells were affected akin the benign MCF10A lines, but to a lesser extent. Thus, GIRK1 regulated cellular pathways in mammary epithelial cells are likely to contribute to the development and progression of breast cancer.


Breast Neoplasms/genetics , Carcinogenesis/genetics , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , Neoplasms/genetics , Breast Neoplasms/pathology , Cell Movement/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Lymphatic Metastasis , MCF-7 Cells , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Neoplasms/pathology , Transcriptome/genetics
5.
Sci Rep ; 5: 8364, 2015 Feb 10.
Article En | MEDLINE | ID: mdl-25666479

Mechanical interaction between cells - specifically distortion of tensional homeostasis-emerged as an important aspect of breast cancer genesis and progression. We investigated the biophysical characteristics of mechanosensitive ion channels (MSCs) in the malignant MCF-7 breast cancer cell line. MSCs turned out to be the most abundant ion channel species and could be activated by negative pressure at the outer side of the cell membrane in a saturable manner. Assessing single channel conductance (GΛ) for different monovalent cations revealed an increase in the succession: Li(+) < Na(+) < K(+) ≈Rb(+) ≈ Cs(+). Divalent cations permeated also with the order: Ca(2+) < Ba(2+). Comparison of biophysical properties enabled us to identify MSCs in MCF-7 as ion channels formed by the Piezo1 protein. Using patch clamp technique no functional MSCs were observed in the benign MCF-10A mammary epithelial cell line. Blocking of MSCs by GsMTx-4 resulted in decreased motility of MCF-7, but not of MCF-10A cells, underscoring a possible role of Piezo1 in invasion and metastatic propagation. The role of Piezo1 in biology and progression of breast cancer is further substantiated by markedly reduced overall survival in patients with increased Piezo1 mRNA levels in the primary tumor.


Breast Neoplasms/metabolism , Cell Movement , Ion Channels/metabolism , Mechanotransduction, Cellular , Neoplasm Proteins/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , Humans , Ion Channels/genetics , Ion Transport/genetics , MCF-7 Cells , Neoplasm Invasiveness , Neoplasm Proteins/genetics
6.
Biochim Biophys Acta ; 1828(4): 1214-21, 2013 Apr.
Article En | MEDLINE | ID: mdl-23305758

G-protein activated inwardly rectifying K(+) channels (GIRKs) of the heterotetrameric GIRK1/GIRK4 composition mediate I(K+ACh) in atrium and are regulated by cAMP dependent protein kinase (PKA). Phosphorylation of GIRK1/GIRK4 complexes promotes the activation of the channel by the G-protein Gßγ-dimer ("heterologous facilitation"). Previously we reported that 3 serines/threonines (S/Ts) within the GIRK1 subunit are phosphorylated by the catalytic subunit of PKA (PKA-cs) in-vitro and are responsible for the acute functional effects exerted by PKA on the homooligomeric GIRK1(F137S) (GIRK1(⁎)) channel. Here we report that homooligomeric GIRK4(WT) and GIRK4(S143T) (GIRK4(⁎)) channels are clearly regulated by PKA phosphorylation. Heterooligomeric channels of the GIRK1(S385CS401CT407C)/GIRK4(WT) composition, where the GIRK1 subunit is devoid of PKA mediated phosphorylation, exhibited reduced but still significant acute effects (reduction during agonist application was ≈49% compared to GIRK1(WT)/GIRK4(WT)). Site directed mutagenesis of truncated cytosolic regions of GIRK4 revealed four serines/threonines (S/Ts) that were heavily phosphorylated by PKA-cs in vitro. Two of them were found to be responsible for the acute effects exerted by PKA in vivo, since the effect of cAMP injection was reduced by ≈99% in homooligomeric GIRK4(⁎T199CS412C) channels. Coexpression of GIRK1(WT)/GIRK4(T199CS412C) reduced the acute effect by ≈65%. Only channels of the GIRK1(S385CS401CT407C)/GIRK4(T199CS412C) composition were practically devoid of PKA mediated effects (reduction by ≈97%), indicating that both subunits contribute to the heterologous facilitation of I(K+ACh).


Cyclic AMP-Dependent Protein Kinases/physiology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/chemistry , Amino Acid Sequence , Animals , Molecular Sequence Data , Phosphorylation , Protein Multimerization , Protein Subunits , Xenopus laevis
7.
PLoS One ; 6(10): e26524, 2011.
Article En | MEDLINE | ID: mdl-22053194

Four-and-a-half LIM domain protein 1 isoform A (FHL1A) is predominantly expressed in skeletal and cardiac muscle. Mutations in the FHL1 gene are causative for several types of hereditary myopathies including X-linked myopathy with postural muscle atrophy (XMPMA). We here studied myoblasts from XMPMA patients. We found that functional FHL1A protein is completely absent in patient myoblasts. In parallel, expression of FHL1C is either unaffected or increased. Furthermore, a decreased proliferation rate of XMPMA myoblasts compared to controls was observed but an increased number of XMPMA myoblasts was found in the G(0)/G(1) phase. Furthermore, low expression of K(v1.5), a voltage-gated potassium channel known to alter myoblast proliferation during the G(1) phase and to control repolarization of action potential, was detected. In order to substantiate a possible relation between K(v1.5) and FHL1C, a pull-down assay was performed. A physical and direct interaction of both proteins was observed in vitro. In addition, confocal microscopy revealed substantial colocalization of FHL1C and K(v1.5) within atrial cells, supporting a possible interaction between both proteins in vivo. Two-electrode voltage clamp experiments demonstrated that coexpression of K(v1.5) with FHL1C in Xenopus laevis oocytes markedly reduced K(+) currents when compared to oocytes expressing K(v1.5) only. We here present the first evidence on a biological relevance of FHL1C.


Intracellular Signaling Peptides and Proteins/metabolism , Kv1.5 Potassium Channel/metabolism , LIM Domain Proteins/metabolism , Muscle Proteins/metabolism , Animals , Blotting, Western , Case-Control Studies , Cell Cycle , Cell Line , Cell Proliferation , Genetic Diseases, X-Linked/metabolism , Genetic Diseases, X-Linked/pathology , Humans , Intracellular Signaling Peptides and Proteins/genetics , Ion Channel Gating , Kinetics , Kv1.5 Potassium Channel/genetics , LIM Domain Proteins/genetics , Male , Mice , Muscle Proteins/genetics , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Mutant Proteins/metabolism , Myoblasts/metabolism , Myoblasts/pathology , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Transport , Reverse Transcriptase Polymerase Chain Reaction , Subcellular Fractions/metabolism , Xenopus , Xenopus laevis
8.
J Cell Biochem ; 110(3): 598-608, 2010 Jun 01.
Article En | MEDLINE | ID: mdl-20512921

The aim of this study was to investigate the impact of increased mRNA levels encoding GIRK1 in breast tumours on GIRK protein expression. mRNA levels encoding hGIRK1 and hGIRK4 in the MCF7, MCF10A and MDA-MB-453 breast cancer cell lines were assessed and the corresponding proteins detected using Western blots. cDNAs encoding for four hGIRK1 splice variants (hGIRK1a, 1c, 1d and 1e) were cloned from the MCF7 cell line. Subcellular localisation of fluorescence labelled hGIRK1a-e and hGIRK4 and of endogenous GIRK1 and GIRK4 subunits was monitored in the MCF7 cell line. All hGIRK1 splice variants and hGIRK4 were predominantly located within the endoplasmic reticulum. Heterologous expression in Xenopus laevis oocytes and two electrode voltage clamp experiments together with confocal microscopy were performed. Only the hGIRK1a subunit was able to form functional GIRK channels in connection with hGIRK4. The other splice variants are expressed, but exert a dominant negative effect on heterooligomeric channel function. Hence, alternative splicing of the KCNJ3 gene transcript in the MCF7 cell line leads to a family of mRNA's, encoding truncated versions of the hGIRK1 protein. The very high abundance of mRNA's encoding GIRK1 together with the presence of GIRK1 protein suggests a pathophysiological role in breast cancer.


Breast Neoplasms/genetics , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , RNA Editing/genetics , Animals , Base Sequence , Blotting, Western , Breast Neoplasms/metabolism , Cell Line, Tumor , Clone Cells , Female , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Humans , Immunohistochemistry , Microscopy, Confocal , Molecular Sequence Data , Patch-Clamp Techniques , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/analysis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Xenopus laevis
9.
FEBS J ; 276(21): 6218-26, 2009 Nov.
Article En | MEDLINE | ID: mdl-19765080

Besides being activated by G-protein beta/gamma subunits, G-protein activated potassium channels (GIRKs) are regulated by cAMP-dependent protein kinase. Back-phosphorylation experiments have revealed that the GIRK1 subunit is phosphorylated in vivo upon protein kinase A activation in Xenopus oocytes, whereas phosphorylation was eliminated when protein kinase A was blocked. In vitro phosphorylation experiments using truncated versions of GIRK1 revealed that the structural determinant is located within the distant, unique cytosolic C-terminus of GIRK1. Serine 385, serine 401 and threonine 407 were identified to be responsible for the incorporation of radioactive (32)P into the protein. Furthermore, the functional effects of cAMP injections into oocytes on currents produced by GIRK1 homooligomers were significantly reduced when these three amino acids were mutated. The data obtained in the present study provide information about the structural determinants that are responsible for protein kinase A phosphorylation and the regulation of GIRK channels.


Cyclic AMP-Dependent Protein Kinases/physiology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Amino Acid Sequence , Animals , Cyclic AMP-Dependent Protein Kinases/chemistry , Female , Molecular Sequence Data , Phosphorylation , Xenopus laevis
...