Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 381
Filter
1.
J Exp Clin Cancer Res ; 42(1): 199, 2023 Aug 09.
Article in English | MEDLINE | ID: mdl-37553567

ABSTRACT

BACKGROUND: Gemcitabine resistance (GR) is a significant clinical challenge in pancreatic adenocarcinoma (PAAD) treatment. Macrophages in the tumor immune-microenvironment are closely related to GR. Uncovering the macrophage-induced GR mechanism could help devise a novel strategy to improve gemcitabine treatment outcomes in PAAD. Therefore, preclinical models accurately replicating patient tumor properties are essential for cancer research and drug development. Patient-derived organoids (PDOs) represent a promising in vitro model for investigating tumor targets, accelerating drug development, and enabling personalized treatment strategies to improve patient outcomes. METHODS: To investigate the effects of macrophage stimulation on GR, co-cultures were set up using PDOs from three PAAD patients with macrophages. To identify signaling factors between macrophages and pancreatic cancer cells (PCCs), a 97-target cytokine array and the TCGA-GTEx database were utilized. The analysis revealed CCL5 and AREG as potential candidates. The role of CCL5 in inducing GR was further investigated using clinical data and tumor sections obtained from 48 PAAD patients over three years, inhibitors, and short hairpin RNA (shRNA). Furthermore, single-cell sequencing data from the GEO database were analyzed to explore the crosstalk between PCCs and macrophages. To overcome GR, inhibitors targeting the macrophage-CCL5-Sp1-AREG feedback loop were evaluated in cell lines, PDOs, and orthotopic mouse models of pancreatic carcinoma. RESULTS: The macrophage-CCL5-Sp1-AREG feedback loop between macrophages and PCCs is responsible for GR. Macrophage-derived CCL5 activates the CCR5/AKT/Sp1/CD44 axis to confer stemness and chemoresistance to PCCs. PCC-derived AREG promotes CCL5 secretion in macrophages through the Hippo-YAP pathway. By targeting the feedback loop, mithramycin improves the outcome of gemcitabine treatment in PAAD. The results from the PDO model were corroborated with cell lines, mouse models, and clinical data. CONCLUSIONS: Our study highlights that the PDO model is a superior choice for preclinical research and precision medicine. The macrophage-CCL5-Sp1-AREG feedback loop confers stemness to PCCs to facilitate gemcitabine resistance by activating the CCR5/AKT/SP1/CD44 pathway. The combination of gemcitabine and mithramycin shows potential as a therapeutic strategy for treating PAAD in cell lines, PDOs, and mouse models.


Subject(s)
Adenocarcinoma , Pancreatic Neoplasms , Animals , Mice , Gemcitabine , Pancreatic Neoplasms/metabolism , Deoxycytidine/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Coculture Techniques , Adenocarcinoma/pathology , Plicamycin/metabolism , Plicamycin/pharmacology , Plicamycin/therapeutic use , Drug Resistance, Neoplasm/genetics , Cell Line, Tumor , Macrophages/metabolism , RNA, Small Interfering/pharmacology , Organoids/metabolism , Tumor Microenvironment , Pancreatic Neoplasms
2.
Biochem Pharmacol ; 197: 114894, 2022 03.
Article in English | MEDLINE | ID: mdl-34968486

ABSTRACT

Mithramycin A (MIT) has reacquired extensive research attention due to its anti-solid tumor activity and improved pharmacological production. Mechanismly, MIT was broadly used as a c-Myc inhibitor, and c-Myc regulated CD47 and PD-L1 expression which has been demonstrated. However, how MIT affects immune check-point molecules remains unknown. In this study, we found CD47 expression was higher in melanoma of pan-tissue array. MIT inhibited CD47 expression both in mRNA and protein level in melanoma cells (SK-MEL-28 and B16). MIT inhibited c-Myc, Sp-1 and CD47 expression in a concentration-dependent way. MIT inhibited the surface CD47 expression and promoted the phagocytosis of SK-MEL-28 cells by THP-1 cells. We found MIT inhibited tumor growth in melanoma allograft mice and CD47 expression in tumor mass. We also found MIT upregulated PD-L1 expression in cancer cells possibly via inhibiting PD-L1 ubiquitination, increasing ROS and IFN-γ. Combination of MIT and anti-PD-1 antibody showed enhanced antitumor activity compared to MIT and anti-PD-1 antibody alone in MC38 allograft mice. Using immune checkpoint array we found MIT inhibited expression of FasL and Galectin3. These results suggest that MIT inhibits CD47 expression, while improves PD-L1 expression. Furthermore, the combination of MIT and anti-PD-1 antibody exerts potent antitumor effect.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , B7-H1 Antigen/biosynthesis , CD47 Antigen/biosynthesis , Melanoma, Experimental/metabolism , Plicamycin/therapeutic use , Animals , Antibiotics, Antineoplastic/pharmacology , B7-H1 Antigen/antagonists & inhibitors , CD47 Antigen/antagonists & inhibitors , Dose-Response Relationship, Drug , Female , Gene Expression , Humans , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Plicamycin/pharmacology , THP-1 Cells , Xenograft Model Antitumor Assays/methods
3.
Cell Death Dis ; 12(11): 978, 2021 10 21.
Article in English | MEDLINE | ID: mdl-34675191

ABSTRACT

Colorectal cancers (CRC) can be classified into four consensus molecular subtypes (CMS), among which CMS1 has the best prognosis, contrasting with CMS4 that has the worst outcome. CMS4 CRC is notoriously resistant against therapeutic interventions, as demonstrated by preclinical studies and retrospective clinical observations. Here, we report the finding that two clinically employed agents, everolimus (EVE) and plicamycin (PLI), efficiently target the prototypic CMS4 cell line MDST8. As compared to the prototypic CMS1 cell line LoVo, MDST8 cells treated with EVE or PLI demonstrated stronger cytostatic and cytotoxic effects, increased signs of apoptosis and autophagy, as well as a more pronounced inhibition of DNA-to-RNA transcription and RNA-to-protein translation. Moreover, nontoxic doses of EVE and PLI induced the shrinkage of MDST8 tumors in mice, yet had only minor tumor growth-reducing effects on LoVo tumors. Altogether, these results suggest that EVE and PLI should be evaluated for their clinical activity against CMS4 CRC.


Subject(s)
Adaptor Proteins, Signal Transducing/drug effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , Cytoskeletal Proteins/drug effects , Everolimus/therapeutic use , Plicamycin/therapeutic use , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Proliferation , Colorectal Neoplasms/pathology , Everolimus/pharmacology , Humans , Mice , Plicamycin/pharmacology
4.
J Nanobiotechnology ; 19(1): 267, 2021 Sep 06.
Article in English | MEDLINE | ID: mdl-34488783

ABSTRACT

BACKGROUND: Sarcomas comprise a group of aggressive malignancies with very little treatment options beyond standard chemotherapy. Reposition of approved drugs represents an attractive approach to identify effective therapeutic compounds. One example is mithramycin (MTM), a natural antibiotic which has demonstrated a strong antitumour activity in several tumour types, including sarcomas. However, its widespread use in the clinic was limited by its poor toxicity profile. RESULTS: In order to improve the therapeutic index of MTM, we have loaded MTM into newly developed nanocarrier formulations. First, polylactide (PLA) polymeric nanoparticles (NPs) were generated by nanoprecipitation. Also, liposomes (LIP) were prepared by ethanol injection and evaporation solvent method. Finally, MTM-loaded hydrogels (HG) were obtained by passive loading using a urea derivative non-peptidic hydrogelator. MTM-loaded NPs and LIP display optimal hydrodynamic radii between 80 and 105 nm with a very low polydispersity index (PdI) and encapsulation efficiencies (EE) of 92 and 30%, respectively. All formulations show a high stability and different release rates ranging from a fast release in HG (100% after 30 min) to more sustained release from NPs (100% after 24 h) and LIP (40% after 48 h). In vitro assays confirmed that all assayed MTM formulations retain the cytotoxic, anti-invasive and anti-stemness potential of free MTM in models of myxoid liposarcoma, undifferentiated pleomorphic sarcoma and chondrosarcoma. In addition, whole genome transcriptomic analysis evidenced the ability of MTM, both free and encapsulated, to act as a multi-repressor of several tumour-promoting pathways at once. Importantly, the treatment of mice bearing sarcoma xenografts showed that encapsulated MTM exhibited enhanced therapeutic effects and was better tolerated than free MTM. CONCLUSIONS: Overall, these novel formulations may represent an efficient and safer MTM-delivering alternative for sarcoma treatment.


Subject(s)
Plicamycin/analogs & derivatives , Plicamycin/pharmacology , Plicamycin/therapeutic use , Sarcoma/pathology , Animals , Anti-Bacterial Agents/therapeutic use , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Chondrosarcoma/drug therapy , Drug Compounding , Female , Humans , Hydrogels/chemistry , Hydrogels/therapeutic use , Liposomes , Mice , Mice, Nude , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Polyesters/chemistry , Polyesters/therapeutic use , Sarcoma/drug therapy
5.
Cell Death Dis ; 11(7): 587, 2020 07 27.
Article in English | MEDLINE | ID: mdl-32719328

ABSTRACT

DNA damage triggers cell death mechanisms contributing to neuronal loss and cognitive decline in neurological disorders, including traumatic brain injury (TBI), and as a side effect of chemotherapy. Mithramycin, which competitively targets chromatin-binding sites of specificity protein 1 (Sp1), was used to examine previously unexplored neuronal cell death regulatory mechanisms via rat primary neurons in vitro and after TBI in mice (males). In primary neurons exposed to DNA-damage-inducing chemotherapy drugs in vitro we showed that DNA breaks sequentially initiate DNA-damage responses, including phosphorylation of ATM, H2AX and tumor protein 53 (p53), transcriptional activation of pro-apoptotic BH3-only proteins, and mitochondrial outer membrane permeabilization (MOMP), activating caspase-dependent and caspase-independent intrinsic apoptosis. Mithramycin was highly neuroprotective in DNA-damage-dependent neuronal cell death, inhibiting chemotherapeutic-induced cell death cascades downstream of ATM and p53 phosphorylation/activation but upstream of p53-induced expression of pro-apoptotic molecules. Mithramycin reduced neuronal upregulation of BH3-only proteins and mitochondrial dysfunction, attenuated caspase-3/7 activation and caspase substrates' cleavage, and limited c-Jun activation. Chromatin immunoprecipitation indicated that mithramycin attenuates Sp1 binding to pro-apoptotic gene promoters without altering p53 binding suggesting it acts by removing cofactors required for p53 transactivation. In contrast, the DNA-damage-independent neuronal death models displayed caspase initiation in the absence of p53/BH3 activation and were not protected even when mithramycin reduced caspase activation. Interestingly, experimental TBI triggers a multiplicity of neuronal death mechanisms. Although markers of DNA-damage/p53-dependent intrinsic apoptosis are detected acutely in the injured cortex and are attenuated by mithramycin, these processes may play a reduced role in early neuronal death after TBI, as caspase-dependent mechanisms are repressed in mature neurons while other, mithramycin-resistant mechanisms are active. Our data suggest that Sp1 is required for p53-mediated transactivation of neuronal pro-apoptotic molecules and that mithramycin may attenuate neuronal cell death in conditions predominantly involving DNA-damage-induced p53-dependent intrinsic apoptosis.


Subject(s)
DNA Damage , Neurons/pathology , Plicamycin/pharmacology , Animals , Apoptosis/drug effects , Biomarkers/metabolism , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/pathology , Cell Death/drug effects , Etoposide/pharmacology , Male , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Models, Biological , Neurons/drug effects , Neuroprotective Agents/pharmacology , Plicamycin/therapeutic use , Proto-Oncogene Proteins c-jun/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Tumor Suppressor Protein p53/metabolism
6.
Article in English | MEDLINE | ID: mdl-31707263

ABSTRACT

The free-living amebae Naegleria, Acanthamoeba, and Balamuthia cause rare but life-threatening infections. All three parasites can cause meningoencephalitis. Acanthamoeba can also cause chronic keratitis and both Balamuthia and Acanthamoeba can cause skin and systemic infections. There are minimal drug development pipelines for these pathogens despite a lack of available treatment regimens and high fatality rates. To identify anti-amebic drugs, we screened 159 compounds from a high-value repurposed library against trophozoites of the three amebae. Our efforts identified 38 compounds with activity against at least one ameba. Multiple drugs that bind the ATP-binding pocket of mTOR and PI3K are active, highlighting these compounds as important inhibitors of these parasites. Importantly, 24 active compounds have progressed at least to phase II clinical studies and overall 15 compounds were active against all three amebae. Based on central nervous system (CNS) penetration or exceptional potency against one amebic species, we identified sixteen priority compounds for the treatment of meningoencephalitis caused by these pathogens. The top five compounds are (i) plicamycin, active against all three free-living amebae and previously U.S. Food and Drug Administration (FDA) approved, (ii) TG02, active against all three amebae, (iii and iv) FDA-approved panobinostat and FDA orphan drug lestaurtinib, both highly potent against Naegleria, and (v) GDC-0084, a CNS penetrant mTOR inhibitor, active against at least two of the three amebae. These results set the stage for further investigation of these clinically advanced compounds for treatment of infections caused by the free-living amebae, including treatment of the highly fatal meningoencephalitis.


Subject(s)
Acanthamoeba/drug effects , Amebiasis/drug therapy , Amoebozoa/drug effects , Antiprotozoal Agents/pharmacology , Central Nervous System Protozoal Infections/drug therapy , Naegleria/drug effects , Amebiasis/parasitology , Carbazoles/pharmacology , Carbazoles/therapeutic use , Cell Culture Techniques , Central Nervous System Protozoal Infections/parasitology , Culture Media , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Furans , Heterocyclic Compounds, 4 or More Rings/pharmacology , Heterocyclic Compounds, 4 or More Rings/therapeutic use , Inhibitory Concentration 50 , Oxazines/pharmacology , Oxazines/therapeutic use , Panobinostat/pharmacology , Panobinostat/therapeutic use , Plicamycin/pharmacology , Plicamycin/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use
7.
Sci Rep ; 9(1): 15202, 2019 10 23.
Article in English | MEDLINE | ID: mdl-31645574

ABSTRACT

The pivotal role of cancer initiating stem cells (CSCs) in tumor initiation, growth, metastasis and drug resistance has led to the postulation of a 'total cancer therapy' paradigm, which involves targeting both cancer cells and CSCs for effective therapy. However, the progress in identifying drugs for total cancer therapy has been limited. Herein, we show for the first time that mithramycin A (Mit-A) can successfully inhibit CSC proliferation, in addition to inhibiting bulk cancer cells in a model of colorectal cancer (CRC), the second leading cause of death among men and women in the United States. To this end, a polymeric nanofiber scaffold culture system was established to develop 3D tumor organoids (tumoroids) from CRC cell lines such as HT29, HCT116, KM12, CT26 and MC38 as well as ex vivo mouse tumors. These tumoroids possessed increased expression of CSC markers and transcription factors, expanded the number of CSCs in culture and increased CSC functional properties measured by aldehyde dehydrogenase activity. Screening of an NCI library of FDA approved drugs led to the identification of Mit-A as a potential total cancer therapy drug. In both sphere and tumoroid culture, Mit-A inhibits cancer growth by reducing the expression of cancer stemness markers. In addition, Mit-A inhibits the expression of SP1, a previously known target in CRCs. Moreover, Mit-A significantly reduces growth of tumoroids in ex vivo cultures and CRC tumor growth in vivo. Finally, a dose-dependent treatment on CRC cells indicate that Mit-A significantly induces the cell death and PARP-cleavage of both CSC and non-CSC cells. Taken together the results of these in vitro, ex vivo and in vivo studies lead to the inference that Mit-A is a promising drug candidate for total cancer therapy of CRCs.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Plicamycin/analogs & derivatives , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Colorectal Neoplasms/pathology , HCT116 Cells , HT29 Cells , Humans , Mice, Inbred C57BL , Neoplastic Stem Cells/pathology , Plicamycin/pharmacology , Plicamycin/therapeutic use
8.
Eur Urol Oncol ; 2(4): 415-424, 2019 07.
Article in English | MEDLINE | ID: mdl-31277777

ABSTRACT

BACKGROUND: The TMPRSS2-ERG gene fusion is the most frequent genetic rearrangement in prostate cancers and results in broad transcriptional reprogramming and major phenotypic changes. Interaction and cooperation of ERG and SP1 may be instrumental in sustaining the tumorigenic and metastatic phenotype and could represent a potential vulnerability in ERG fusion-positive tumors. OBJECTIVE: To test the activity of EC-8042, a compound able to block SP1, in cellular and mouse models of ERG-positive prostate cancer. DESIGN, SETTING, AND PARTICIPANTS: We evaluated the activity of EC-8042 in cell cultures and ERG/PTEN transgenic/knockout mice that provide reliable models for testing novel therapeutics in this specific disease context. Using a new protocol to generate tumor spheroids from ERG/PTEN mice, we also examined the effects of EC-8042 on tumor-propagating stem-like cancer cells with high self-renewal and tumorigenic capabilities. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: The efficacy of EC-8042 was determined by measuring the proliferative capacity and target gene expression in cell cultures, invasive and metastatic capabilities in chick chorioallantoic membrane assays, and tumor development in mice. Significance was determined using statistical test. RESULTS AND LIMITATIONS: EC-8042 blocked transcription of ERG-regulated genes and reverted the invasive and metastatic phenotype of VCaP cells. EC-8042 blocked the expansion of stem-like tumor cells in tumor spheroids from VCaP cells and mouse-derived tumors. In ERG/PTEN mice, systemic treatment with EC-8042 inhibited ERG-regulated gene transcription, tumor progression, and tumor-propagating stem-like tumor cells. CONCLUSIONS: Our data support clinical testing of EC-8042 for the treatment of ERG-positive prostate cancer in precision medicine approaches. PATIENT SUMMARY: In this study, EC-8042, a novel compound with a favorable pharmacological and toxicological profile, exhibited relevant activity in cell cultures and in vivo in a genetically engineered mouse model that closely recapitulates the features of clinically aggressive ERG-positive prostate cancer. Our data indicate that further evaluation of EC-8042 in clinical trials is warranted.


Subject(s)
Plicamycin/analogs & derivatives , Prostatic Neoplasms/genetics , Sp1 Transcription Factor/antagonists & inhibitors , Transcriptional Regulator ERG/genetics , Animals , Cell Line, Tumor , Humans , Male , Mice, Transgenic , Neoplastic Stem Cells , PTEN Phosphohydrolase/genetics , Plicamycin/pharmacology , Plicamycin/therapeutic use , Prostatic Neoplasms/drug therapy
9.
Mol Oncol ; 12(9): 1498-1512, 2018 09.
Article in English | MEDLINE | ID: mdl-29738634

ABSTRACT

Chemoresistance is a major therapeutic challenge that plays a role in the poor statistical outcomes in pancreatic cancer. Unfolded protein response (UPR) is one of the homeostasis mechanisms in cancer cells that have been correlated with chemoresistance in a number of cancers including pancreatic cancer. In this study, we show that modulating glucose regulatory protein 78 (GRP78), the master regulator of the UPR, can have a profound effect on multiple pathways that mediate chemoresistance. Our study showed for the first time that silencing GRP78 can diminish efflux activity of ATP-binding cassette (ABC) transporters, and it can decrease the antioxidant response resulting in an accumulation of reactive oxygen species (ROS). We also show that these effects can be mediated by the activity of specificity protein 1 (SP1), a transcription factor overexpressed in pancreatic cancer. Thus, inhibition of SP1 negatively affects the UPR, deregulates the antioxidant response of NRF2, as well as ABC transporter activity by inhibiting GRP78-mediated ER homeostasis. Sp1 and NRF2 have been classified as nononcogene addiction genes and thus are imperative to understanding the molecular mechanism of resistance. These finding have huge clinical relevance as both Sp1 and GRP78 are overexpressed in pancreatic cancer patients and increased expression of these proteins is indicative of poor prognosis. Understanding how these proteins may regulate chemoresistance phenotype of this aggressive cancer may pave the way for development of efficacious therapy for this devastating disease.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Antibiotics, Antineoplastic/therapeutic use , Drug Resistance, Neoplasm , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Plicamycin/therapeutic use , Reactive Oxygen Species/metabolism , Animals , Antibiotics, Antineoplastic/pharmacology , Cell Death/drug effects , Cell Line, Tumor , Disease Progression , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Gene Expression , Gene Silencing , Homeostasis , Humans , Mice , Mice, Nude , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Pancreatic Neoplasms/pathology , Plicamycin/pharmacology , Sp1 Transcription Factor/antagonists & inhibitors , Sp1 Transcription Factor/metabolism , Unfolded Protein Response
10.
Int J Mol Sci ; 19(5)2018 May 09.
Article in English | MEDLINE | ID: mdl-29747385

ABSTRACT

Osteoarthritis (OA) is the most common and increasing joint disease worldwide. Current treatment for OA is limited to control of symptoms. The purpose of this study was to determine the effect of specificity protein 1 (SP1) inhibitor Mithramycin A (MitA) on chondrocyte catabolism and OA pathogenesis and to explore the underlying molecular mechanisms involving SP1 and other key factors that are critical for OA. Here, we show that MitA markedly inhibited expressions of matrix-degrading enzymes induced by pro-inflammatory cytokine interleukin-1β (IL-1β) in mouse primary chondrocytes. Intra-articular injection of MitA into mouse knee joint alleviated OA cartilage destruction induced by surgical destabilization of the medial meniscus (DMM). However, modulation of SP1 level in chondrocyte and mouse cartilage did not alter catabolic gene expression or cartilage integrity, respectively. Instead, MitA significantly impaired the expression of HIF-2α known to be critical for OA pathogenesis. Such reduction in expression of HIF-2α by MitA was caused by inhibition of NF-κB activation, at least in part. These results suggest that MitA can alleviate OA pathogenesis by suppressing NF-κB-HIF-2α pathway, thus providing insight into therapeutic strategy for OA.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Osteoarthritis/drug therapy , Plicamycin/analogs & derivatives , Animals , Cartilage, Articular/drug effects , Cells, Cultured , Chondrocytes/drug effects , Chondrocytes/enzymology , Chondrocytes/metabolism , Disease Progression , Enzyme Induction/drug effects , Interleukin-1beta/pharmacology , Joints/pathology , Male , Matrix Metalloproteinases/metabolism , Mice, Inbred C57BL , NF-kappa B/metabolism , Osteoarthritis/enzymology , Osteoarthritis/pathology , Plicamycin/administration & dosage , Plicamycin/pharmacology , Plicamycin/therapeutic use , Sp1 Transcription Factor/metabolism
11.
Exp Neurol ; 302: 145-154, 2018 04.
Article in English | MEDLINE | ID: mdl-29337144

ABSTRACT

The progressive loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) is one of the hallmarks of Parkinson's disease (PD). Neuroinflammation has been proposed to contributes to the progressive nature of the disease. Early growth response-1 (Egr-1), a zinc finger transcription factor, has been shown to have a crucial role in both neuronal death and the inflammatory response. However, whether and how Egr-1 is involved in the pathogenesis of PD has not been investigated. Using the subacute 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD, we identified early peak induction of Egr-1 in the SNpc but not in the striatum. In situ immunofluorescent analysis showed that Egr-1 predominantly locates in the nuclei of nigral AldoC (+) astrocytes upon MPTP treatment. Genetic ablation of Egr-1 or inhibition of its transcriptional activity by Mithramycin A significantly suppresses the activation of both astrocytes and microglia, decreases proinflammatory cytokine expression, and protects dopaminergic cell bodies from degeneration in the SNpc. Taken together, these findings demonstrate that the induction of Egr-1 promotes neuroinflammation and dopaminergic cell body loss in the SNpc of MPTP-induced mouse model, suggesting an important role of astrocytic Egr-1 in neuroinflammation in PD.


Subject(s)
Dopamine/metabolism , Early Growth Response Protein 1/metabolism , Inflammation/etiology , Inflammation/pathology , Neurodegenerative Diseases/etiology , Parkinsonian Disorders/complications , Animals , Astrocytes/metabolism , Astrocytes/pathology , CD11b Antigen/genetics , CD11b Antigen/metabolism , Calcium-Binding Proteins/metabolism , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Early Growth Response Protein 1/genetics , Enzyme Inhibitors/therapeutic use , Male , Mice , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Neurodegenerative Diseases/drug therapy , Parkinsonian Disorders/drug therapy , Phosphopyruvate Hydratase/metabolism , Plicamycin/analogs & derivatives , Plicamycin/therapeutic use , Substantia Nigra/metabolism , Time Factors , Tyrosine 3-Monooxygenase/metabolism
12.
Cancer Chemother Pharmacol ; 80(3): 645-652, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28735378

ABSTRACT

PURPOSE: In a preclinical drug screen, mithramycin was identified as a potent inhibitor of the Ewing sarcoma EWS-FLI1 transcription factor. We conducted a phase I/II trial to determine the dose-limiting toxicities (DLT), maximum tolerated dose (MTD), and pharmacokinetics (PK) of mithramycin in children with refractory solid tumors, and the activity in children and adults with refractory Ewing sarcoma. PATIENTS AND METHODS: Mithramycin was administered intravenously over 6 h once daily for 7 days for 28 day cycles. Adult patients (phase II) initially received mithramycin at the previously determined recommended dose of 25 µg/kg/dose. The planned starting dose for children (phase I) was 17.5 µg/kg/dose. Plasma samples were obtained for mithramycin PK analysis. RESULTS: The first two adult patients experienced reversible grade 4 alanine aminotransferase (ALT)/aspartate aminotransferase (AST) elevation exceeding the MTD. Subsequent adult patients received mithramycin at 17.5 µg/kg/dose, and children at 13 µg/kg/dose with dexamethasone pretreatment. None of the four subsequent adult and two pediatric patients experienced cycle 1 DLT. No clinical responses were observed. The average maximal mithramycin plasma concentration in four patients was 17.8 ± 4.6 ng/mL. This is substantially below the sustained mithramycin concentrations ≥50 nmol/L required to suppress EWS-FLI1 transcriptional activity in preclinical studies. Due to inability to safely achieve the desired mithramycin exposure, the trial was closed to enrollment. CONCLUSIONS: Hepatotoxicity precluded the administration of a mithramycin at a dose required to inhibit EWS-FLI1. Evaluation of mithramycin in patients selected for decreased susceptibility to elevated transaminases may allow for improved drug exposure.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Oncogene Proteins, Fusion/metabolism , Plicamycin/therapeutic use , Proto-Oncogene Protein c-fli-1/metabolism , RNA-Binding Protein EWS/metabolism , Sarcoma, Ewing/drug therapy , Adolescent , Adult , Antibiotics, Antineoplastic/pharmacokinetics , Child , Female , Humans , Male , Plicamycin/pharmacokinetics , Sarcoma, Ewing/pathology , Young Adult
13.
Arch Dermatol Res ; 309(8): 611-623, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28695331

ABSTRACT

SAHA (vorinostat) is a histone deacetylase inhibitor approved by the USA Food and Drug Administration (FDA) for treating advanced refractory cutaneous T cell lymphomas. As SAHA alters the expression of many genes under control of the Sp1 transcription factor, we examined the effect of its association with the FDA-approved anticancer antibiotic Mithramycin A (MTR, plicamycin), a competitive inhibitor of Sp1 binding to DNA. Sézary syndrome (SS) cells, expanded ex vivo from peripheral blood mononuclear cells of 4 patients, were tested for their sensitivity to the drugs regarding cytotoxicity and differential responsive gene expression. Multivariate statistical methods were used to identify genes whose expression is altered by SAHA, MTR, and the synergist effect of the two drugs. MTR, like SAHA, induced the apoptosis of SS cells, while the two drugs in combination showed clear synergy or potentiation. Expression data stressed a likely important role of additive or synergistic epigenetic modifications in the combined effect of the two drugs, while direct inhibition of Sp1-dependent transcription seemed to have only limited impact. Ontological analysis of modified gene expression suggested that the two drugs, either independently or synergistically, counteracted many intertwined pro-survival pathways deregulated in SS cells, resistance of these tumors to intrinsic and extrinsic apoptosis, abnormal adhesion migration, and invasive properties, as well as immunosuppressive behavior. Our findings provide preliminary clues on the individual and combined effects of SAHA and MTR in SS cells and highlight a potential therapeutic interest of this novel pair of drugs for treatment of SS patients.


Subject(s)
Hydroxamic Acids/therapeutic use , Plicamycin/therapeutic use , Sezary Syndrome/drug therapy , Skin Neoplasms/drug therapy , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Drug Therapy, Combination , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hydroxamic Acids/administration & dosage , Plicamycin/administration & dosage , Transcriptome , Vorinostat
14.
Neurochem Res ; 41(8): 1924-38, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27072684

ABSTRACT

Increasing evidence has shown that specificity protein 1 (Sp1) is abnormally increased in the brains of subjects with Alzheimer's disease (AD) and transgenic AD models. However, whether the Sp1 activation plays a critical role in the AD pathogenesis and selective inhibition of Sp1 activation may have a disease-modifying effect on the AD-like phenotypes remain elusive. In this study, we reported that Sp1 mRNA and protein expression were markedly increased in the brain of APPswe/PS1dE9 transgenic mice, whereas chronic administration of mithramycin A (MTM), a selective Sp1 inhibitor, potently inhibited Sp1 activation in the APPswe/PS1dE9 mice down to the levels of wild-type mice. Specifically, we found that MTM treatment resulted in a significant improvement of learning and memory deficits, a dramatic reduction in cerebral Aß levels and plaque burden, a profound reduction in tau hyperphosphorylation, and a marked increase in synaptic marker in the APPswe/PS1dE9 mice. In addition, MTM treatment was powerfully effective in inhibiting amyloid precursor protein (APP) processing via suppressing APP, beta-site APP cleaving enzyme 1 (BACE1), and presenilin-1 (PS1) mRNA and protein expression to preclude Aß production in the APPswe/PS1dE9 mice. Furthermore, MTM treatment strongly inhibited phosphorylated CDK5 and GSK3ß signal pathways to reduce tau hyperphosphorylation in the APPswe/PS1dE9 mice. Collectively, our findings provide evidence that Sp1 activation may contribute to the AD pathogenesis and may serve as a novel therapeutic target in the treatment of AD. The present study highlights that selective Sp1 inhibitors may be considered as disease-modifying therapeutic agents for AD.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Cognition Disorders/drug therapy , Cognition Disorders/pathology , Disease Models, Animal , Plicamycin/analogs & derivatives , Alzheimer Disease/metabolism , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cognition Disorders/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Male , Mice , Mice, Transgenic , Plicamycin/pharmacology , Plicamycin/therapeutic use , Sp1 Transcription Factor/antagonists & inhibitors , Sp1 Transcription Factor/metabolism
15.
Oncotarget ; 7(21): 30935-50, 2016 May 24.
Article in English | MEDLINE | ID: mdl-27105533

ABSTRACT

Tumor initiating cells (TICs), responsible for tumor initiation, and cancer stem cells (CSCs), responsible for tumor expansion and propagation, are often resistant to chemotherapeutic agents. To find therapeutic targets against sarcoma initiating and propagating cells we used models of myxoid liposarcoma (MLS) and undifferentiated pleomorphic sarcoma (UPS) developed from human mesenchymal stromal/stem cells (hMSCs), which constitute the most likely cell-of-origin for sarcoma. We found that SP1-mediated transcription was among the most significantly altered signaling. To inhibit SP1 activity, we used EC-8042, a mithramycin (MTM) analog (mithralog) with enhanced anti-tumor activity and highly improved safety. EC-8042 inhibited the growth of TIC cultures, induced cell cycle arrest and apoptosis and upregulated the adipogenic factor CEBPα. SP1 knockdown was able to mimic the anti-proliferative effects induced by EC-8042. Importantly, EC-8042 was not recognized as a substrate by several ABC efflux pumps involved in drug resistance, and, opposite to the chemotherapeutic drug doxorubicin, repressed the expression of many genes responsible for the TIC/CSC phenotype, including SOX2, C-MYC, NOTCH1 and NFκB1. Accordingly, EC-8042, but not doxorubicin, efficiently reduced the survival of CSC-enriched tumorsphere sarcoma cultures. In vivo, EC-8042 induced a profound inhibition of tumor growth associated to a strong reduction of the mitotic index and the induction of adipogenic differentiation and senescence. Finally, EC-8042 reduced the ability of tumor cells to reinitiate tumor growth. These data suggest that EC-8042 could constitute an effective treatment against both TIC and CSC subpopulations in sarcoma.


Subject(s)
Antineoplastic Agents/therapeutic use , Gene Expression Regulation, Neoplastic/drug effects , Neoplastic Stem Cells/drug effects , Plicamycin/analogs & derivatives , Sarcoma, Experimental/drug therapy , Sp1 Transcription Factor/metabolism , ATP-Binding Cassette Transporters/metabolism , Animals , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Doxorubicin/pharmacokinetics , Drug Resistance, Neoplasm , Female , Fluorescent Antibody Technique , Humans , Mice, Inbred NOD , Mice, SCID , NF-kappa B p50 Subunit/metabolism , Plicamycin/pharmacokinetics , Plicamycin/therapeutic use , Proto-Oncogene Proteins c-myc/metabolism , Receptor, Notch1/metabolism , SOXB1 Transcription Factors/metabolism , Sarcoma, Experimental/genetics , Sarcoma, Experimental/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
16.
J Am Anim Hosp Assoc ; 51(5): 291-9, 2015.
Article in English | MEDLINE | ID: mdl-26355578

ABSTRACT

Hypercalcemia is uncommonly encountered in veterinary patients. When it does occur, the effects can be severe, resulting in significant morbidity and mortality if not recognized and addressed in a timely manner. Causes of hypercalcemia are varied and include pituitary-dependent and pituitary-independent causes. A diagnosis of hypercalcemia should be made based on documentation of ionized hypercalcemia. The mainstay of emergency treatment usually involves aggressive IV fluid diuresis, the use of diuretics, and, often, glucocorticoids. The use of bisphosphonates has become increasingly more common in veterinary medicine.


Subject(s)
Calcium/metabolism , Cat Diseases/diagnosis , Diphosphonates/therapeutic use , Dog Diseases/diagnosis , Hypercalcemia/veterinary , Animals , Calcimimetic Agents/therapeutic use , Calcium Chelating Agents/therapeutic use , Cat Diseases/etiology , Cat Diseases/therapy , Cats , Diuretics/therapeutic use , Dog Diseases/etiology , Dog Diseases/therapy , Dogs , Edetic Acid/therapeutic use , Fluid Therapy/veterinary , Gallium/therapeutic use , Homeostasis/physiology , Hypercalcemia/diagnosis , Hypercalcemia/etiology , Hypercalcemia/therapy , Plicamycin/therapeutic use
17.
Planta Med ; 81(15): 1326-38, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26393942

ABSTRACT

Mithramycin is an antitumor compound of the aureolic acid family produced by Streptomyces argillaceus. It has been used to treat several types of cancer including testicular carcinoma, chronic and acute myeloid leukemia as well as hypercalcemias and Paget's disease. Although the use of mithramycin in humans has been limited because its side effects, in recent years a renewed interest has arisen since new uses and activities have been ascribed to it. Chemically, mithramycin is characterized by a tricyclic aglycone bearing two aliphatic side chains attached at C3 and C7, and disaccharide and trisaccharide units attached at positions 2 and 6, respectively. The mithramycin gene cluster has been characterized. This has allowed for the development of several mithramycin analogs ("mithralogs") by combinatorial biosynthesis and/or biocatalysis. The combinatorial biosynthesis strategies include gene inactivation and/or the use of sugar biosynthesis plasmids for sugar modification. In addition, lipase-based biocatalysis enabled selective modifications of the hydroxyl groups, providing further mithramycin analogs. As a result, new mithramycin analogs with higher antitumor activity and/or less toxicity have been generated. One, demycarosyl-3D-ß-D-digitoxosyl-mithramycin SK (EC-8042), is being tested in regulatory preclinical assays, representing an opportunity to open the therapeutic window of this promising molecular scaffold.


Subject(s)
Antibiotics, Antineoplastic/chemistry , Plicamycin/analogs & derivatives , Animals , Antibiotics, Antineoplastic/therapeutic use , Biocatalysis , Combinatorial Chemistry Techniques , Humans , Plicamycin/chemistry , Plicamycin/therapeutic use , Streptomyces/chemistry
18.
Transl Res ; 165(5): 558-77, 2015 May.
Article in English | MEDLINE | ID: mdl-25468484

ABSTRACT

Chemotherapy resistance is a major clinical challenge for the management of locally advanced breast cancer. Accumulating evidence suggests a major role of cancer stem cells (CSCs) in chemoresistance evoking the requirement of drugs that selectively target CSCs in combination with chemotherapy. Here, we report that mithramycin A, a known specificity protein (Sp)1 inhibitor, sensitizes breast CSCs (bCSCs) by perturbing the expression of drug efflux transporters, ATP-binding cassette sub-family G, member 2 (ABCG2) and ATP-binding cassette sub-family C, member 1 (ABCC1), survival factors, B-cell lymphoma 2 (Bcl-2) and X-linked inhibitor of apoptosis (XIAP), and, stemness regulators, octamer-binding transcription factor 4 (Oct4) and Nanog, which are inherently upregulated in these cells compared with the rest of the tumor population. In-depth analysis revealed that aberrant overexpression of Sp1 in bCSCs transcriptionally upregulates (1) resistance-promoting genes to protect these cells from genotoxic therapy, and (2) stemness regulators to sustain self-renewal potential of these cells. However, mithramycin A causes transcriptional suppression of these chemoresistant and self-renewal genes by inhibiting Sp1 recruitment to their promoters. Under such antisurvival microenvironment, chemotherapeutic agent doxorubicin induces apoptosis in bCSCs via DNA damage-induced reactive oxygen species generation. Cumulatively, our findings raise the possibility that mithramycin A might emerge as a promising drug in combinatorial therapy with the existing chemotherapeutic agents that fail to eliminate CSCs. This will consequently lead to the improvement of therapeutic outcome for the treatment-resistant breast carcinomas.


Subject(s)
Breast Neoplasms/drug therapy , Doxorubicin/therapeutic use , Neoplastic Stem Cells/drug effects , Plicamycin/analogs & derivatives , Adult , Aged , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols , Apoptosis/drug effects , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Drug Resistance, Neoplasm/drug effects , Female , Humans , MCF-7 Cells , Middle Aged , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Plicamycin/therapeutic use , Sp1 Transcription Factor/antagonists & inhibitors , Sp1 Transcription Factor/genetics , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Translational Research, Biomedical , Tumor Cells, Cultured , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics
19.
Sci Rep ; 4: 7162, 2014 Nov 24.
Article in English | MEDLINE | ID: mdl-25418289

ABSTRACT

Cervical cancer is the third most common cancer and the third leading cause of death among women. However, the standard treatment for cervical cancer includes cisplatin, which can cause side effects such as hematological damage or renal toxicity. New innovations in cervical cancer treatment focus on developing more effective and better-tolerated therapies such as Sp1-targeting drugs. Previous studies suggested that mithramycin A (Mith) inhibits the growth of various cancers by decreasing Sp1 protein. However, how Sp1 protein is decreased by Mith is not clear. Few studies have investigated the regulation of Sp1 protein by proteasome-dependent degradation as a possible control mechanism for the regulation of Sp1 in cancer cells. Here, we show that Mith decreased Sp1 protein by inducing proteasome-dependent degradation, thereby suppressing cervical cancer growth through a DR5/caspase-8/Bid signaling pathway. We found that prolonged Mith treatment was well tolerated after systemic administration to mice carrying cervical cancer cells. Reduction of body weight was minimal, indicating that Mith was a good therapeutic candidate for treatment of cancers in which Sp1 is involved in promoting and developing disease.


Subject(s)
Cell Proliferation/drug effects , Plicamycin/analogs & derivatives , Signal Transduction/drug effects , Sp1 Transcription Factor/metabolism , Uterine Cervical Neoplasms/drug therapy , Animals , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/therapeutic use , Apoptosis/drug effects , BH3 Interacting Domain Death Agonist Protein/metabolism , Caspase 8/metabolism , Cell Line, Tumor , Female , Humans , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Nude , Mitochondria/drug effects , Mitochondria/metabolism , Peptide Hydrolases/metabolism , Plicamycin/pharmacology , Plicamycin/therapeutic use , RNA Interference , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Sp1 Transcription Factor/antagonists & inhibitors , Sp1 Transcription Factor/genetics , Transplantation, Heterologous , Uterine Cervical Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...