24 research outputs found

    Pacjent z podejrzeniem zespołu pozornego nadmiaru mineralokortykosteroidów — trudności diagnostyczne

    Get PDF
    Pozorny nadmiar mineralokortykosteroidów (AME) jest rzadką postacią dziedzicznego nadciśnienia tętniczego związaną z zaburzeniami w funkcjonowaniu dehydrogenazy 11β-hydroksysteroidowej 2. Diagnostyka tego schorzenia opiera się na ocenie fenotypu, na który składają się charakterystyczne objawy kliniczne, zaburzenia w gospodarce elektrolitowej oraz analiza profilu steroidowego (mineralo- i glikokortykosteroidowego). Ponadto wykonuje się genotypowanie mające na celu ustalenie występowania mutacji w genie HSD11B2. W niniejszej pracy przedstawiono trudności w diagnostyce AME na podstawie przypadku pacjenta, u którego ocena jedynie fenotypu mogłaby doprowadzić do błędnego rozpoznania

    Sodium Butyrate Enhances Curcuminoids Permeability through the Blood-Brain Barrier, Restores Wnt/β-Catenin Pathway Antagonists Gene Expression and Reduces the Viability of Glioblastoma Cells

    No full text
    Glioblastoma (GBM) is an extremely aggressive brain tumor awaiting novel, efficient, and minimally toxic treatment. Curcuminoids (CCM), polyphenols from Curcuma longa, and sodium butyrate (NaBu), a histone deacetylase inhibitor naturally occurring in the human body, await elucidation as potential anti-GBM agents. Thus, the aim of this study was to analyze CCM and NaBu both separately and as a combination treatment using three GBM cell lines. MTT was used for cytotoxicity evaluation, and the combination index was calculated for synergism prediction. Cell cycle, apoptosis, and reactive oxygen species (ROS) generation were analyzed using flow cytometry. DNA methylation was verified by MS-HRM and mRNA expression by qPCR. The permeability through the blood-brain barrier (BBB) and through the nasal cavity was evaluated using PAMPA model. The results of this study indicate that CCM and NaBu synergistically reduce the viability of GBM cells inducing apoptosis and cell cycle arrest. These effects are mediated via ROS generation and changes in gene expression, including upregulation of Wnt/β-catenin pathway antagonists, SFRP1, and RUNX3, and downregulation of UHRF1, the key epigenetic regulator. Moreover, NaBu ameliorated CCM permeability through the BBB and the nasal cavity. We conclude that CCM and NaBu are promising agents with anti-GBM properties

    Novel Approaches to Epigenetic Therapies: From Drug Combinations to Epigenetic Editing

    No full text
    Cancer development involves both genetic and epigenetic alterations. Aberrant epigenetic modifications are reversible, allowing excellent opportunities for therapeutic intervention. Nowadays, several epigenetic drugs are used worldwide to treat, e.g., myelodysplastic syndromes and leukemias. However, overcoming resistance and widening the therapeutic profiles are the most important challenges faced by traditional epigenetic drugs. Recently, novel approaches to epigenetic therapies have been proposed. Next-generation epigenetic drugs, with longer half-life and better bioavailability, are being developed and tested. Since epigenetic phenomena are interdependent, treatment modalities include co-administration of two different epigenetic drugs. In order to sensitize cancer cells to chemotherapy, epigenetic drugs are administered prior to chemotherapy, or both epigenetic drug and chemotherapy are used together to achieve synergistic effects and maximize treatment efficacy. The combinations of epigenetic drug with immunotherapy are being tested, because they have proved to enhance antitumor immune responses. The next approach involves targeting the metabolic causes of epigenetic changes, i.e., enzymes which, when mutated, produce oncometabolites. Finally, epigenome editing makes it possible to modify individual chromatin marks at a defined region with unprecedented specificity and efficiency. This review summarizes the above attempts in fulfilling the promise of epigenetic drugs in the effective cancer treatment

    Novel Approaches to Epigenetic Therapies: From Drug Combinations to Epigenetic Editing

    No full text
    Cancer development involves both genetic and epigenetic alterations. Aberrant epigenetic modifications are reversible, allowing excellent opportunities for therapeutic intervention. Nowadays, several epigenetic drugs are used worldwide to treat, e.g., myelodysplastic syndromes and leukemias. However, overcoming resistance and widening the therapeutic profiles are the most important challenges faced by traditional epigenetic drugs. Recently, novel approaches to epigenetic therapies have been proposed. Next-generation epigenetic drugs, with longer half-life and better bioavailability, are being developed and tested. Since epigenetic phenomena are interdependent, treatment modalities include co-administration of two different epigenetic drugs. In order to sensitize cancer cells to chemotherapy, epigenetic drugs are administered prior to chemotherapy, or both epigenetic drug and chemotherapy are used together to achieve synergistic effects and maximize treatment efficacy. The combinations of epigenetic drug with immunotherapy are being tested, because they have proved to enhance antitumor immune responses. The next approach involves targeting the metabolic causes of epigenetic changes, i.e., enzymes which, when mutated, produce oncometabolites. Finally, epigenome editing makes it possible to modify individual chromatin marks at a defined region with unprecedented specificity and efficiency. This review summarizes the above attempts in fulfilling the promise of epigenetic drugs in the effective cancer treatment

    Spinal muscular atrophy - onasemnogene abeparvovec and other therapeutic options

    No full text
    Spinal muscular atrophy (SMA) is a neuromuscular disorder that results in the loss of motor neurons. SMA is caused by mutations in the SMN1 gene, leading to the decreased synthesis of the SMN protein, necessary for motor neuron survival. In the past, SMA was considered to be an incurable disease, and therapy was limited only to symptomatic treatment. However, currently there are drugs which effectively inhibit the development of the disease, and even give hope for its cure. Their mechanism of action involves either delivering of a functional copy of the SMN1 gene, or modification of the alternative splicing of the SMN2 gene. Moreover, amelioration of muscle growth and increasing muscle contractility serves as a way of relieving the symptoms of the disease. The functional copy of SMN1 gene is delivered by onasemnogene abeparvovec (Zolgensma). The drug contains cDNA sequences, which correspond to the missing SMN1 gene. It is administered in the form of adeno-associated viral vector serotype 9 (AAV9)–based gene therapy, as a single intravenous infusion, to treat children less than 2 years old. Currently, the drug is approved only in the USA, and its cost exceeds PLN 2,000,000. SMN2 has nearly identical sequence as SMN1, however due to alternative splicing, only around 10% of its transcript results in a full-length, functional SMN protein. Modification of the alternative splicing of the SMN2 pre-mRNA by the drug nusinersen (Spinraza) results in an increased level of the SMN protein. Nusinersen is administered as intrathecal injections and is available both in the USA as well as in Europe. Also risdiplam and branaplam - small-molecule drugs with similar mechanism of action are now being tested in clinical trials. The inhibition proMyostatin cleavage and slowing calcium release, leads to the increased muscle growth and contractility, respectively. So far, three promising drugs are being evaluated in clinical trials: SRK-015, which is a selective and local inhibitor of the activation of myostatin, as well as reldesemtiv (CK-2127107) and tirasemtiv (CK-2127107), both being the fast skeletal muscle troponin activators. These drugs do not affect the genetic cause of SMA, but relieve the symptoms of the disease. Early diagnosis and treatment gives hope for halting the progress of SMA, preserving motor function and extending patient’s life.Rdzeniowy zanik mięśni (SMA) jest neurodegeneracyjną chorobą genetyczną, charakteryzującą się obumieraniem neuronów ruchowych. Przyczyną SMA są mutacje w genie SMN1, które skutkują niedoborem białka SMN. Niegdyś SMA uważane było za chorobę nieuleczalną, a terapia sprowadzała się do leczenia objawowego. Jednak obecnie dostępne są leki, które skutecznie hamują rozwój choroby, a wręcz dają nadzieję na jej wyleczenie. Ich mechanizm działania polega na dostarczeniu funkcjonalnej kopii genu SMN1, lub na zmodyfikowaniu składania transkryptu SMN2. Funkcjonalną kopię genu SMN1 dostarcza preparat onasemnogene abeparvovec (Zolgensma). Lek zawiera sekwencje cDNA, które odpowiadają prawidłowemu genowi SMN1 i podawany jest z wykorzystaniem kapsydu wirusa AAV9 jako nośnika w pojedynczej infuzji dożylnej. Leczeniem mogą być objęte dzieci do 2 roku życia. Obecnie lek zarejestrowany jest jedynie w USA, zaś koszt terapii przekracza 2 mln złotych. Gen SMN2 posiada sekwencję niemal identyczną z genem SMN1, jednak w wyniku alternatywnego składania pierwotnego transkryptu mRNA tego genu powstaje tylko ok. 10% funkcjonalnego białka SMN. Modyfikacja składania transkryptu genu SMN2 przez preparat nusinersen (Spinraza), zwiększa poziom funkcjonalnego białka SMN u leczonych nim chorych. Preparat podawany jest dooponowo. Leczenie możliwe jest zarówno w USA jak i na terenie Europy. Ponadto, testowane są również drobnocząsteczkowe leki o podobnym mechanizmie działania – risdiplam oraz branaplam. Stymulacja wzrostu mięśni oraz nasilenie ich kurczliwości stanowi również element terapii łagodzącej objawy choroby. Obecnie w fazie badań są trzy obiecujące związki: SRK-015, będący inhibitorem miostatyny, oraz reldesemtiv (CK-2127107) i tirasemtiv (CK-2127107), które są szybkimi aktywatorami troponiny. Wczesna diagnoza i podjęcie leczenia daje szansę na zahamowanie rozwoju SMA i tym samym utrzymanie sprawności motorycznej pacjenta oraz przedłużenie jego życia

    ABCB1 Is Frequently Methylated in Higher-Grade Gliomas and May Serve as a Diagnostic Biomarker of More Aggressive Tumors

    No full text
    ABCB1 belongs to a superfamily of membrane transporters that use ATP hydrolysis to efflux various endogenous compounds and drugs outside the cell. Cancer cells upregulate ABCB1 expression as an adaptive response to evade chemotherapy-mediated cell death. On the other hand, several reports highlight the role of the epigenetic regulation of ABCB1 expression. In fact, the promoter methylation of ABCB1 was found to be methylated in several tumor types, including gliomas, but its role as a biomarker is not fully established yet. Thus, the aim of this study was to analyze the methylation of the ABCB1 promoter in tumor tissues from 50 glioma patients to verify its incidence and to semi-quantitively detect ABCB1 methylation levels in order to establish its utility as a potential biomarker. The results of this study show a high interindividual variability in the ABCB1 methylation level of the samples derived from gliomas of different grades. Additionally, a positive correlation between ABCB1 methylation, the WHO tumor grade, and an IDH1 wild-type status has been observed. Thus, ABCB1 methylation can be regarded as a potential diagnostic or prognostic biomarker for glioma patients, indicating more aggressive tumors

    Cannabidiol and Its Combinations with Nonsteroidal Anti-Inflammatory Drugs Induce Apoptosis and Inhibit Activation of NF-κB Signaling in Vulvar Squamous Cell Carcinoma

    No full text
    Vulvar squamous cell carcinoma (VSCC) is a rare malignancy with a relatively good prognosis. However, the prognosis remains poor for elderly patients and those with a significant depth of tumor invasion; thus, novel treatment modalities are needed. The aim of this study was to analyze the impact of cannabidiol (CBD) and its combination with NSAIDs, diclofenac (DIC) and ibuprofen (IBU) on VSCC cells. In this regard, the MTT test was applied for cytotoxicity analysis. Moreover, the influence of CBD, DIC and IBU, as well as their combinations, on apoptosis and cell cycle distribution were analyzed by flow cytometry. The mechanisms of action of the analyzed compounds, including their impact on NF-κB signaling, p53 and COX-2 expression were evaluated using Western blot. This study shows that CBD and its combinations with NSAIDs are cytotoxic to A431 cells, but they also reduce, in a dose-dependent manner, the viability of immortalized keratinocyte HaCaT cells, and human umbilical vein cell line, EA.hy926. Moreover, the compounds and their combinations induced apoptosis, diminished the NF-κB signaling activation and reduced COX-2 expression. We conclude that CBD and its combination with DIC or IBU are promising candidates for the adjuvant treatment of high-risk VSCC patients. However, their impact on non-cancerous cells requires careful evaluation

    Honokiol-Loaded Nanoemulsion for Glioblastoma Treatment: Statistical Optimization, Physicochemical Characterization, and an In Vitro Toxicity Assay

    No full text
    Background: Glioblastoma (GBM) is an extremely invasive and heterogenous malignant brain tumor. Despite advances in current anticancer therapy, treatment options for glioblastoma remain limited, and tumor recurrence is inevitable. Therefore, alternative therapies or new active compounds that can be used as adjuvant therapy are needed. This study aimed to develop, optimize, and characterize honokiol-loaded nanoemulsions intended for intravenous administration in glioblastoma therapy. Methods: Honokiol-loaded nanoemulsion was developed by incorporating honokiol into Lipofundin MCT/LCT 20% using a horizontal shaker. The Box–Behnken design, coupled with response surface methodology, was used to optimize the incorporation process. The effect of the developed formulation on glioblastoma cell viability was determined using the MTT test. Long-term and short-term stress tests were performed to evaluate the effect of honokiol on the stability of the oil-in-water system and the effect of different stress factors on the stability of honokiol, respectively. Its physicochemical properties, such as MDD, PDI, ZP, OSM, pH, and loading efficiency (LE%), were determined. Results: The optimized honokiol-loaded nanoemulsion was characterized by an MDD of 201.4 (0.7) nm with a PDI of 0.07 (0.02) and a ZP of −28.5 (0.9) mV. The LE% of honokiol was above 95%, and pH and OSM were sufficient for intravenous administration. The developed formulation was characterized by good stability and a satisfactory toxicity effect of the glioblastoma cell lines. Conclusions: The honokiol-loaded nanoemulsion is a promising pharmaceutical formulation for further development in the adjuvant therapy of glioblastoma

    Honokiol-Loaded Nanoemulsion for Glioblastoma Treatment: Statistical Optimization, Physicochemical Characterization, and an In Vitro Toxicity Assay

    No full text
    Background: Glioblastoma (GBM) is an extremely invasive and heterogenous malignant brain tumor. Despite advances in current anticancer therapy, treatment options for glioblastoma remain limited, and tumor recurrence is inevitable. Therefore, alternative therapies or new active compounds that can be used as adjuvant therapy are needed. This study aimed to develop, optimize, and characterize honokiol-loaded nanoemulsions intended for intravenous administration in glioblastoma therapy. Methods: Honokiol-loaded nanoemulsion was developed by incorporating honokiol into Lipofundin MCT/LCT 20% using a horizontal shaker. The Box–Behnken design, coupled with response surface methodology, was used to optimize the incorporation process. The effect of the developed formulation on glioblastoma cell viability was determined using the MTT test. Long-term and short-term stress tests were performed to evaluate the effect of honokiol on the stability of the oil-in-water system and the effect of different stress factors on the stability of honokiol, respectively. Its physicochemical properties, such as MDD, PDI, ZP, OSM, pH, and loading efficiency (LE%), were determined. Results: The optimized honokiol-loaded nanoemulsion was characterized by an MDD of 201.4 (0.7) nm with a PDI of 0.07 (0.02) and a ZP of −28.5 (0.9) mV. The LE% of honokiol was above 95%, and pH and OSM were sufficient for intravenous administration. The developed formulation was characterized by good stability and a satisfactory toxicity effect of the glioblastoma cell lines. Conclusions: The honokiol-loaded nanoemulsion is a promising pharmaceutical formulation for further development in the adjuvant therapy of glioblastoma
    corecore