1
|
Peng J, Wei CI, Lee SH. Eeyarestatin I (ESI)-induced ERAD inhibition exhibits anti-cancer activity through multiple mechanisms in human colorectal cancer cells. Eur J Pharmacol 2025; 997:177623. [PMID: 40222444 DOI: 10.1016/j.ejphar.2025.177623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 04/10/2025] [Accepted: 04/11/2025] [Indexed: 04/15/2025]
Abstract
Endoplasmic reticulum (ER)-associated degradation (ERAD) is a cellular process for maintenance of protein homeostasis in the ER and aberration of ERAD regulation leads to abnormal function of ER. As an inhibitory compound to ERAD, Eeyarestatin I (ESI) exhibits anti-cancer activity. In this study, we elucidated the anti-cancer mechanisms of ESI-induced ERAD inhibition in human colorectal carcinoma cells. Cellular viability of three different types of human colorectal cancer cells decreased in a dose-dependent manner by treatment with ESI. Treatment of ESI to human colorectal cancer cells led to significant increase of ubiquitin accumulation, G2/M phase cell cycle arrest, apoptosis, ER stress and autophagy. In addition, ESI treatment reduced transcriptional activity of nuclear factor kappa B (NF-κB), and increased phosphorylation of c-Jun NH2-terminal kinase (JNK) and intracellular production of reactive oxygen species (ROS). Decrease of cell viability and ROS release were JNK-dependent and apoptosis was ROS-dependent. On the other hand, treatment of the cells with ESI downregulated the expression of translocon-associated protein (TRAP) subunits including TRAPα, β, γ and δ, which was JNK- and ROS-dependent. In summary, ESI-induced ERAD inhibition triggers ER stress, G2/M cell cycle arrest, ROS-dependent apoptosis, and autophagy in human colorectal cancer cells. We are the first to identify TRAPs as novel target ER membrane proteins that are downregulated by ERAD inhibition in human colorectal cancer cells.
Collapse
Affiliation(s)
- Jing Peng
- Department of Nutrition and Food Science, College of Agriculture and Natural Resources, University of Maryland College Park, College Park, MD, 20742, USA
| | - Cheng-I Wei
- Department of Nutrition and Food Science, College of Agriculture and Natural Resources, University of Maryland College Park, College Park, MD, 20742, USA
| | - Seong-Ho Lee
- Department of Nutrition and Food Science, College of Agriculture and Natural Resources, University of Maryland College Park, College Park, MD, 20742, USA.
| |
Collapse
|
2
|
Lauro G, Aliberti M, De Nisco M, Pedatella S, Pepe G, Basilicata MG, Chini MG, Fischer K, Hofstetter RK, Werz O, Ferraro MG, Piccolo M, Irace C, Saviano A, Campiglia P, Bertamino A, Ostacolo C, Ciaglia T, Manfra M, Bifulco G. Furazanopyrazine-based novel promising anticancer agents interfering with the eicosanoid biosynthesis pathways by dual mPGES-1 and sEH inhibition. Eur J Med Chem 2025; 289:117402. [PMID: 40010271 DOI: 10.1016/j.ejmech.2025.117402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 02/28/2025]
Abstract
We report the identification of a new set of compounds based on the furazanopyrazine core interfering with eicosanoid biosynthesis and acting as potentially effective anti-inflammatory and anticancer agents. Based on our previous promising results on a set of furazanopyrazine-based compounds against the microsomal prostaglandin E2 synthase-1 (mPGES-1) enzyme, we here identified derivatives with improved pharmacokinetic properties by replacing the ester moiety with a more stable ether group. A focused virtual library of 1 × 104 molecules was built and screened against mPGES-1 through molecular docking experiments, leading to the selection of 10 candidates for synthesis and biological evaluation. Several molecules were found to inhibit mPGES-1 and, among them, two items featured IC50 values in the low micromolar range. Additional computational studies on the collection of synthesized compounds demonstrated that compound 3b, previously emerged as an mPGES-1 inhibitor, interfered with soluble epoxide hydrolase (sEH) activity, thus emerging as a valuable dual mPGES-1/sEH inhibitor. The pharmacokinetic features of the most potent compounds were accurately estimated. Unfortunately, poor outcomes were obtained for 3b; on the other hand, compound 7e exhibited promising mPGES-1 inhibition and excellent pharmacokinetic profile, demonstrating that the novel furazanopyrazine-based items with ether moiety possess improved pharmacokinetic properties compared to the ester-based compounds reported in our previous study. Additionally, the anticancer properties of 7e and 7d, the latter emerged as the most active mPGES-1 inhibitor, were evaluated and both compounds showed promising activities against HCT-116 human colorectal cancer (CRC) cells. These findings highlight the furazanopyrazine core as a promising scaffold for disclosing new anti-inflammatory drugs with the ability to inhibit targets belonging to arachidonic acid cascade.
Collapse
Affiliation(s)
- Gianluigi Lauro
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, Fisciano, 84084, Italy
| | - Michela Aliberti
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, Fisciano, 84084, Italy
| | - Mauro De Nisco
- Department of Health Sciences, University of Basilicata, Viale dell'Ateneo Lucano, Potenza, I-85100, Italy
| | - Silvana Pedatella
- Department of Chemical Sciences, University of Napoli Federico II, Via Cintia 4, I-80126, Napoli, Italy
| | - Giacomo Pepe
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, Fisciano, 84084, Italy
| | - Manuela Giovanna Basilicata
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", P.zza L. Miraglia 2, 80138, Naples, Italy
| | - Maria Giovanna Chini
- Department of Biosciences and Territory, University of Molise, C.da Fonte Lappone, Pesche, 86090, Italy
| | - Katrin Fischer
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, Jena, 07743, Germany
| | - Robert K Hofstetter
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, Jena, 07743, Germany
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, Jena, 07743, Germany
| | - Maria Grazia Ferraro
- Department of Molecular Medicine and Medical Biotechnologies, School of Medicine and Surgery, University of Naples, Via Domenico Montesano 49, Naples, 80131, Italy
| | - Marialuisa Piccolo
- BioChem Lab, Department of Pharmacy, School of Medicine and Surgery, University of Naples, Via Domenico Montesano 49, Naples, 80131, Italy
| | - Carlo Irace
- BioChem Lab, Department of Pharmacy, School of Medicine and Surgery, University of Naples, Via Domenico Montesano 49, Naples, 80131, Italy
| | - Anella Saviano
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, Fisciano, 84084, Italy
| | - Alessia Bertamino
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, Fisciano, 84084, Italy
| | - Carmine Ostacolo
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, Fisciano, 84084, Italy
| | - Tania Ciaglia
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, Fisciano, 84084, Italy
| | - Michele Manfra
- Department of Health Sciences, University of Basilicata, Viale dell'Ateneo Lucano, Potenza, I-85100, Italy.
| | - Giuseppe Bifulco
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, Fisciano, 84084, Italy.
| |
Collapse
|
3
|
Keikhosravi A, Guin K, Pegoraro G, Misteli T. Simulation and Quantitative Analysis of Spatial Centromere Distribution Patterns. Cells 2025; 14:491. [PMID: 40214445 PMCID: PMC11987964 DOI: 10.3390/cells14070491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/18/2025] [Accepted: 03/22/2025] [Indexed: 04/14/2025] Open
Abstract
A prominent feature of eukaryotic chromosomes are centromeres, which are specialized regions of repetitive DNA required for faithful chromosome segregation during cell division. In interphase cells, centromeres are non-randomly positioned in the three-dimensional space of the nucleus in a cell type-specific manner. The functional relevance and the cellular mechanisms underlying this localization are unknown, and quantitative methods to measure distribution patterns of centromeres in 3D space are needed. Here, we developed an analytical framework that combines sensitive clustering metrics and advanced modeling techniques for the quantitative analysis of centromere distributions at the single-cell level. To identify a robust quantitative measure for centromere clustering, we benchmarked six metrics for their ability to sensitively detect changes in centromere distribution patterns from high-throughput imaging data of human cells, both under normal conditions and upon experimental perturbation of centromere distribution. We found that Ripley's K function has the highest accuracy with minimal sensitivity to variations in the number of centromeres, making it the most suitable metric for measuring centromere distributions. As a complementary approach, we also developed and validated spatial models to replicate centromere distribution patterns, and we show that a radially shifted Gaussian distribution best represents the centromere patterns seen in human cells. Our approach creates tools for the quantitative characterization of spatial centromere distributions with applications in both targeted studies of centromere organization and unbiased screening approaches.
Collapse
Affiliation(s)
- Adib Keikhosravi
- High Throughput Imaging Facility (HiTIF), National Cancer Institute, NIH, Bethesda, MD 20892, USA;
| | - Krishnendu Guin
- Cell Biology of Genomes Group, National Cancer Institute, NIH, Bethesda, MD 20892, USA;
| | - Gianluca Pegoraro
- High Throughput Imaging Facility (HiTIF), National Cancer Institute, NIH, Bethesda, MD 20892, USA;
| | - Tom Misteli
- Cell Biology of Genomes Group, National Cancer Institute, NIH, Bethesda, MD 20892, USA;
| |
Collapse
|
4
|
Huang Z, Whitehead B, Nejsum P, Corredig M, Rasmussen MK. Tomato-derived extracellular vesicles increase intestinal zinc transportation by potentially down-regulating the expression of the metallothionein family. Food Res Int 2025; 203:115804. [PMID: 40022334 DOI: 10.1016/j.foodres.2025.115804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 01/15/2025] [Accepted: 01/18/2025] [Indexed: 03/03/2025]
Abstract
Extracellular vesicles (EVs) have the ability to regulate physiological and pathological processes across species and have been shown to be present in plants. Tomatoes are one of the most widespread vegetables on the market and exhibit a broad range of health-promoting effects, including antioxidant and anti-inflammatory properties. However, little is known about the bioactivity of tomato-derived EVs. Here, we isolated EVs from tomatoes and explored their gene regulatory potential using array-based transcriptomics. Interestingly, using a differentiated Caco-2 monolayer model, tomato-derived EVs were shown to upregulate the transportation of zinc, which may involve the down-regulation of metallothionein proteins (MTs). Differentiated Caco-2 cells internalized tomato-derived EVs. Post-EV treatment the relative expression levels of MT-related mRNAs within the cells decreased by approximately threefold, accompanied by an approximately twofold reduction in intracellular zinc concentration. Additionally, the amount of secreted zinc in the basolateral medium increased by approximately threefold. Moreover, tomato-derived EV regulation of MT gene expression occurred only in differentiated epithelial cells. This effect was observed in differentiated Caco-2 and HIEC-6 cells, whereas no impact was seen on the MT gene in undifferentiated cells. This mechanistic study uniquely demonstrates the bioactivity of tomato-derived EVs, and for the first time, reveals the ability of plant-derived EVs to modify zinc regulation across the intestinal epithelia. This further suggests the potential of plant-derived EVs as functional food supplements in the future.
Collapse
Affiliation(s)
- Ziyu Huang
- Department of Food Science Aarhus University Denmark
| | - Bradley Whitehead
- Department of Clinical Medicine Aarhus University Aarhus Denmark; Department of Infectious Diseases Aarhus University Hospital Aarhus Denmark
| | - Peter Nejsum
- Department of Clinical Medicine Aarhus University Aarhus Denmark; Department of Infectious Diseases Aarhus University Hospital Aarhus Denmark
| | | | | |
Collapse
|
5
|
Keikhosravi A, Guin K, Pegoraro G, Misteli T. Simulation and quantitative analysis of spatial centromere distribution patterns. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.22.634320. [PMID: 39896519 PMCID: PMC11785228 DOI: 10.1101/2025.01.22.634320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
A prominent feature of eukaryotic chromosomes are centromeres, which are specialized regions of repetitive DNA required for faithful chromosome segregation during cell division. In interphase cells centromeres are non-randomly positioned in the three-dimensional space of the nucleus in a cell-type specific manner. The functional relevance and the cellular mechanisms underlying this observation are unknown, and quantitative methods to measure distribution patterns of centromeres in 3D space are needed. Here we have developed an analytical framework that combines robust clustering metrics and advanced modeling techniques for the quantitative analysis of centromere distributions at the single cell level. To identify a robust quantitative measure for centromere clustering, we benchmarked six metrics for their ability to sensitively detect changes in centromere distribution patterns from high-throughput imaging data of human cells, both under normal conditions and upon experimental perturbation of centromere distribution. We find that Ripley's K Score has the highest accuracy with minimal sensitivity to variations in centromeres number, making it the most suitable metric for measuring centromere distributions. As a complementary approach, we also developed and validated spatial models to replicate centromere distribution patterns, and we show that a radially shifted Gaussian distribution best represents the centromere patterns seen in human cells. Our approach creates tools for the quantitative characterization of spatial centromere distributions with applications in both targeted studies of centromere organization as well as in unbiased screening approaches.
Collapse
Affiliation(s)
- Adib Keikhosravi
- High Throughput Imaging Facility (HiTIF), National Cancer Institute, NIH, Bethesda, MD 20892
| | - Krishnendu Guin
- Cell Biology of Genomes Group, National Cancer Institute, NIH, Bethesda, MD 20892
| | - Gianluca Pegoraro
- High Throughput Imaging Facility (HiTIF), National Cancer Institute, NIH, Bethesda, MD 20892
| | - Tom Misteli
- Cell Biology of Genomes Group, National Cancer Institute, NIH, Bethesda, MD 20892
| |
Collapse
|
6
|
Mederle AL, Semenescu A, Drăghici GA, Dehelean CA, Vlăduț NV, Nica DV. Sodium Butyrate: A Multifaceted Modulator in Colorectal Cancer Therapy. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:136. [PMID: 39859117 PMCID: PMC11766496 DOI: 10.3390/medicina61010136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 01/06/2025] [Accepted: 01/13/2025] [Indexed: 01/30/2025]
Abstract
Background and Objectives: Sodium butyrate (NaB) is a potent modulator of cancer-related gene networks. However, its precise mechanisms of action and effects at elevated doses remain insufficiently explored. This study investigated the impact of NaB at physiologically relevant doses on key cellular metrics (viability, confluence, cell number, morphology, nuclear integrity) and a comprehensive set of apoptosis and proliferation regulators (including underexplored genes) in colorectal cancer (CRC) cells. Materials and Methods: Human HCT-116 cells were treated with increasing NaB concentrations (0-20 mM). Cell viability, confluence, number, morphology, and nuclear integrity were assessed using MTT and imaging assays. RT-PCR was used to determine changes in the expression of critical pro-apoptotic players (BAX, CASP3, PUMA, TP53), anti-apoptotic facilitators (BCL-2, MCL-1), cell division regulators (PCNA, Ki-67, CDKN1), and inflammation genes (NF-κB). Results: This study provides the first exploration of MCL-1 and PCNA modulation by NaB in the context of CRC and HCT-116 cells, offering significant translational insights. All treatments reduced cell viability, confluence, and number in a dose-dependent manner (p < 0.0001). Gene expression revealed dose-related increases in most pro-apoptotic markers (BAX, CASP3, PUMA; p < 0.001), and decreases for the other genes (p < 0.001). BAX emerged as the most responsive gene to NaB, while TP53 showed minimal sensitivity, supporting NaB's effectiveness in p53-compromised phenotypes. Nuclear condensation and fragmentation at higher NaB doses confirmed apoptotic induction. Conclusions: NaB can modulate critical apoptotic and cell cycle genes, disrupt tumor cell proliferation, and overcome resistance mechanisms associated with anti-apoptotic regulators such as MCL-1. By targeting both short-term and long-term anti-apoptotic defenses, NaB shows promise as a preventive and therapeutic agent in CRC, particularly in high-risk phenotypes with compromised p53 functionality. These findings support its potential for integration into combination therapies or dietary interventions aimed at enhancing colonic butyrate levels.
Collapse
Affiliation(s)
- Alexandra Laura Mederle
- Doctoral School, “Victor Babeș” University of Medicine and Pharmacy Timişoara, Eftimie Murgu Square No. 2, 300041 Timișoara, Romania;
| | - Alexandra Semenescu
- Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, 300041 Timișoara, Romania; (A.S.); (C.A.D.)
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy,“Victor Babeş” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, 300041 Timișoara, Romania;
| | - George Andrei Drăghici
- Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, 300041 Timișoara, Romania; (A.S.); (C.A.D.)
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy,“Victor Babeş” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, 300041 Timișoara, Romania;
| | - Cristina Adriana Dehelean
- Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, 300041 Timișoara, Romania; (A.S.); (C.A.D.)
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy,“Victor Babeş” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, 300041 Timișoara, Romania;
| | - Nicolae-Valentin Vlăduț
- The National Institute of Research—Development for Machines and Installations Designed for Agriculture and Food Industry (INMA), Bulevardul Ion Ionescu de la Brad 6, 077190 București, Romania;
| | - Dragoş Vasile Nica
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy,“Victor Babeş” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, 300041 Timișoara, Romania;
- The National Institute of Research—Development for Machines and Installations Designed for Agriculture and Food Industry (INMA), Bulevardul Ion Ionescu de la Brad 6, 077190 București, Romania;
| |
Collapse
|
7
|
Colarusso E, Lauro G, Potenza M, Galatello P, Garigliota MLD, Ferraro MG, Piccolo M, Chini MG, Irace C, Campiglia P, Hoffstetter RK, Werz O, Ramunno A, Bifulco G. 5-methyl-2-carboxamidepyrrole-based novel dual mPGES-1/sEH inhibitors as promising anticancer candidates. Arch Pharm (Weinheim) 2025; 358:e2400708. [PMID: 39692230 DOI: 10.1002/ardp.202400708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/19/2024]
Abstract
Inhibiting microsomal prostaglandin E2 synthase-1 (mPGES-1), an inducible enzyme involved in prostaglandin E2 (PGE2) biosynthesis and tumor microenvironment (TME) homeostasis, is a valuable strategy for treating inflammation and cancer. In this work, 5-methylcarboxamidepyrrole-based molecules were designed and synthesized as new compounds targeting mPGES-1. Remarkably, compounds 1f, 2b, 2c, and 2d were able to significantly reduce the activity of the isolated enzyme, showing IC50 values in the low micromolar range. With the aim of further profiling the synthesized molecules, their ability to interfere with the activity of soluble epoxide hydrolase (sEH), whose inhibition blocks the loss of the anti-inflammatory mediators epoxyeicosatrienoic acids (EETs or epoxyicosatrienoic acids), was investigated in silico and by employing specific biological assays. Among the set of tested compounds, 1f, 2b, 2c, and 2d emerged as mPGES-1/sEH dual inhibitors. Moreover, given that overexpression of mPGES-1 has been observed in many human tumors, we finally explored the biological effect of our compounds in an in vitro model of human colorectal cancer (CRC). The obtained outcomes pave the way for future investigation to optimize and further characterize anticancer pharmacological profile of the carboxamidepyrrole-based molecules.
Collapse
Affiliation(s)
- Ester Colarusso
- Department of Pharmacy, University of Salerno, Fisciano, Italy
| | - Gianluigi Lauro
- Department of Pharmacy, University of Salerno, Fisciano, Italy
| | - Marianna Potenza
- Department of Pharmacy, University of Salerno, Fisciano, Italy
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University, Jena, Germany
| | - Paola Galatello
- Department of Pharmacy, University of Salerno, Fisciano, Italy
| | | | - Maria Grazia Ferraro
- Department of Molecular Medicine and Medical Biotechnologies, School of Medicine and Surgery, University of Naples, Naples, Italy
| | - Marialuisa Piccolo
- BioChem Lab, Department of Pharmacy, School of Medicine and Surgery, University of Naples, Naples, Italy
| | | | - Carlo Irace
- BioChem Lab, Department of Pharmacy, School of Medicine and Surgery, University of Naples, Naples, Italy
| | | | - Robert Klaus Hoffstetter
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University, Jena, Germany
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University, Jena, Germany
| | - Anna Ramunno
- Department of Pharmacy, University of Salerno, Fisciano, Italy
| | | |
Collapse
|
8
|
Wilson JJ, Bennie L, Eguaogie O, Elkashif A, Conlon PF, Jena L, McErlean E, Buckley N, Englert K, Dunne NJ, Tucker JHR, Vyle JS, McCarthy HO. Synthesis and characterisation of a nucleotide based pro-drug formulated with a peptide into a nano-chemotherapy for colorectal cancer. J Control Release 2024; 369:63-74. [PMID: 38513729 DOI: 10.1016/j.jconrel.2024.03.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 03/01/2024] [Accepted: 03/18/2024] [Indexed: 03/23/2024]
Abstract
Recent studies in colorectal cancer patients (CRC) have shown that increased resistance to thymidylate synthase (TS) inhibitors such as 5-fluorouracil (5-FU), reduce the efficacy of standard of care (SoC) treatment regimens. The nucleotide pool cleanser dUTPase is highly expressed in CRC and is an attractive target for potentiating anticancer activity of chemotherapy. The purpose of the current work was to investigate the activity of P1, P4-di(2',5'-dideoxy-5'-selenouridinyl)-tetraphosphate (P4-SedU2), a selenium-modified symmetrically capped dinucleoside with prodrug capabilities that is specifically activated by dUTPase. Using mechanochemistry, P4-SedU2 and the corresponding selenothymidine analogue P4-SeT2 were prepared with a yield of 19% and 30% respectively. The phosphate functionality facilitated complexation with the amphipathic cell-penetrating peptide RALA to produce nanoparticles (NPs). These NPs were designed to deliver P4-SedU2 intracellularly and thereby maximise in vivo activity. The NPs demonstrated effective anti-cancer activity and selectivity in the HCT116 CRC cell line, a cell line that overexpresses dUTPase; compared to HT29 CRC cells and NCTC-929 fibroblast cells which have reduced levels of dUTPase expression. In vivo studies in BALB/c SCID mice revealed no significant toxicity with respect to weight or organ histology. Pharmacokinetic analysis of blood serum showed that RALA facilitates effective delivery and rapid internalisation into surrounding tissues with NPs eliciting lower plasma Cmax than the equivalent injection of free P4-SedU2, translating the in vitro findings. Tumour growth delay studies have demonstrated significant inhibition of growth dynamics with the tumour doubling time extended by >2weeks. These studies demonstrate the functionality and action of a new pro-drug nucleotide for CRC.
Collapse
Affiliation(s)
- Jordan J Wilson
- School of Pharmacy, Queen's University Belfast, Medical Biological Centre, 97 Lisburn Road, Belfast BT9 7LB, UK; School of Chemistry and Chemical Engineering, Queen's University Belfast, David Keir Building, Stranmillis Road, Belfast BT9 5AG, UK
| | - Lindsey Bennie
- School of Pharmacy, Queen's University Belfast, Medical Biological Centre, 97 Lisburn Road, Belfast BT9 7LB, UK
| | - Olga Eguaogie
- School of Chemistry and Chemical Engineering, Queen's University Belfast, David Keir Building, Stranmillis Road, Belfast BT9 5AG, UK
| | - Ahmed Elkashif
- School of Pharmacy, Queen's University Belfast, Medical Biological Centre, 97 Lisburn Road, Belfast BT9 7LB, UK
| | - Patrick F Conlon
- School of Chemistry and Chemical Engineering, Queen's University Belfast, David Keir Building, Stranmillis Road, Belfast BT9 5AG, UK
| | - Lynn Jena
- School of Pharmacy, Queen's University Belfast, Medical Biological Centre, 97 Lisburn Road, Belfast BT9 7LB, UK
| | - Emma McErlean
- School of Pharmacy, Queen's University Belfast, Medical Biological Centre, 97 Lisburn Road, Belfast BT9 7LB, UK
| | - Niamh Buckley
- School of Pharmacy, Queen's University Belfast, Medical Biological Centre, 97 Lisburn Road, Belfast BT9 7LB, UK
| | - Klaudia Englert
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Nicholas J Dunne
- School of Mechanical and Manufacturing Engineering, Dublin City University, Centre for Medical Engineering Research, Dublin City University, Ireland
| | - James H R Tucker
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Joseph S Vyle
- School of Chemistry and Chemical Engineering, Queen's University Belfast, David Keir Building, Stranmillis Road, Belfast BT9 5AG, UK
| | - Helen O McCarthy
- School of Pharmacy, Queen's University Belfast, Medical Biological Centre, 97 Lisburn Road, Belfast BT9 7LB, UK; School of Chemical Sciences, Dublin City University, Collins Avenue, Dublin 9, Ireland.
| |
Collapse
|
9
|
Jafari SF, Keshavarzi M, AbdulMajid AM, Al-Suede FSR, Asif M, Ahamed MBK, Khan MSS, Hassan LAE, Majid ASA, Naseri M. Evaluation of in vitro and in vivo anticancer activities of potassium koetjapate: a solubility improved formulation of koetjapic acid against human colon cancer. Res Pharm Sci 2024; 19:203-216. [PMID: 39035582 PMCID: PMC11257210 DOI: 10.4103/rps.rps_247_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 08/21/2023] [Accepted: 03/10/2024] [Indexed: 07/23/2024] Open
Abstract
Background and purpose The previous work on koetjapic acid (KA) isolated from Sandoricum koetjape showed its efficacy towards colorectal cancer however KA has poor water solubility which poses the biggest hindrance to its efficacy. In the present paper, an attempt was made to study the anti-colon cancer efficacy of KA's potassium salt i.e. potassium koetjapate (KKA) applying in vitro and in vivo methods. Experimental approach KKA was produced by a semi-synthetic method. A human apoptosis proteome profiler array was applied to determine the protein targets responsible for the stimulation of apoptosis. Three doses of KKA were studied in athymic nude mice models to examine the in vivo anti-tumorigenic ability of KKA. Findings/Results The results of this study demonstrated that KKA regulates the activities of various proteins. It downregulates the expression of several antiapoptotic proteins and negative regulators of apoptosis including HSP60, HSP90, Bcl-2, and IGF-1 in HCT 116 cells with consequent upregulation of TRAILR-1 and TRAILR-2, p27, CD40, caspase 3, and caspase 8 proteins. Additionally, KKA showed an in vitro antimetastatic effect against HCT 116 cells. These results are feasibly related to the down-regulation of Notch, Wnt, hypoxia, and MAPK/JNK and MAPK/ERK signalling pathways in HCT 116 cells besides the up-regulation of a transcription factor for cell cycle (pRb-E2F) pathways. In addition, KKA revealed potent inhibition of tumor growth. Conclusion and implications In sum, the findings indicate that KKA can be a promising candidate as a chemotherapeutic agent against colorectal cancer.
Collapse
Affiliation(s)
- Seyedeh Fatemeh Jafari
- Department of Plant Sciences, Faculty of Biological Sciences, Alzahra University, Tehran, Iran
| | - Maryam Keshavarzi
- Department of Plant Sciences, Faculty of Biological Sciences, Alzahra University, Tehran, Iran
| | - Amin MalikShah AbdulMajid
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, Australian National University, Australia
- EMAN Research Ltd. level 10-14, Wormald Street, Symonston, ACT 2009, Australia and EMAN Biodiscoveries Sdn. Bhd., A1-4, Halal Park, 08000 Sungai Petani, Kedah, Malaysia. Malaysia
| | - Fouad Saleih R. Al-Suede
- EMAN Research Ltd. level 10-14, Wormald Street, Symonston, ACT 2009, Australia and EMAN Biodiscoveries Sdn. Bhd., A1-4, Halal Park, 08000 Sungai Petani, Kedah, Malaysia. Malaysia
| | - Muhammad Asif
- Department of Pharmacology, Faculty of Pharmacy, The Islamia University of Bahawalpur 63100 Punjab, Pakistan
| | | | | | | | - Aman Shah Abdul Majid
- Department of Pharmacology, Faculty of Medicine, Quest International University, Malaysia
| | - Mohsen Naseri
- Traditional Medicine Clinical Trial Research Centre, Shahed University, Tehran, Iran
| |
Collapse
|
10
|
Parascandolo A, Benincasa G, Corcione F, Laukkanen MO. ERK2 Is a Promoter of Cancer Cell Growth and Migration in Colon Adenocarcinoma. Antioxidants (Basel) 2024; 13:119. [PMID: 38247543 PMCID: PMC10812609 DOI: 10.3390/antiox13010119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/14/2024] [Accepted: 01/16/2024] [Indexed: 01/23/2024] Open
Abstract
ERK1/2 phosphorylation is frequently downregulated in the early phase of colon tumorigenesis with subsequent activation of ERK5. In the current work, we studied the advantages of ERK1/2 downregulation for tumor growth by dissecting the individual functions of ERK1 and ERK2. The patient sample data demonstrated decreased ERK1/2 phosphorylation in the early phase of tumorigenesis followed by increased phosphorylation in late-stage colon adenocarcinomas with intratumoral invasion or metastasis. In vitro results indicated that SOD3-mediated coordination of small GTPase RAS regulatory genes inhibited RAS-ERK1/2 signaling. In vitro and in vivo studies suggested that ERK2 has a more prominent role in chemotactic invasion, collective migration, and cell proliferation than ERK1. Of note, simultaneous ERK1 and ERK2 expression inhibited collective cell migration and proliferation but tended to promote invasion, suggesting that ERK1 controls ERK2 function. According to the present data, phosphorylated ERK1/2 at the early phase of colon adenocarcinoma limits tumor mass expansion, whereas reactivation of the kinases at the later phase of colon carcinogenesis is associated with the initiation of metastasis. Additionally, our results suggest that ERK1 is a regulatory kinase that coordinates ERK2-promoted chemotactic invasion, collective migration, and cell proliferation. Our findings indicate that ROS, especially H2O2, are associated with the regulation of ERK1/2 phosphorylation in colon cancer by either increasing or decreasing kinase activity. These data suggest that ERK2 has a growth-promoting role and ERK1 has a regulatory role in colon tumorigenesis, which could lead to new avenues in the development of cancer therapy.
Collapse
Affiliation(s)
- Alessia Parascandolo
- Department of Translational Medical Sciences, University of Naples Federico II, Via Pansini 5, 80131 Naples, Italy;
| | | | | | - Mikko O. Laukkanen
- Department of Translational Medical Sciences, University of Naples Federico II, Via Pansini 5, 80131 Naples, Italy;
- Center for Experimental Endocrinology and Oncology (IEOS), CNR-IEOS, Via Pansini 5, 80131 Naples, Italy
| |
Collapse
|
11
|
Chitale S, Wu W, Mukherjee A, Lannon H, Suresh P, Nag I, Ambrosi CM, Gertner RS, Melo H, Powers B, Wilkins H, Hinton H, Cheah M, Boynton Z, Alexeyev A, Sword D, Basan M, Park H, Ham D, Abbott J. A semiconductor 96-microplate platform for electrical-imaging based high-throughput phenotypic screening. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.01.543281. [PMID: 37333319 PMCID: PMC10274629 DOI: 10.1101/2023.06.01.543281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Profiling compounds and genetic perturbations via high-content imaging has become increasingly popular for drug discovery, but the technique is limited to endpoint images of fixed cells. In contrast, electronic-based devices offer label-free, functional information of live cells, yet current approaches suffer from low-spatial resolution or single-well throughput. Here, we report a semiconductor 96-microplate platform designed for high-resolution real-time impedance "imaging" at scale. Each well features 4,096 electrodes at 25 µm spatial resolution while a miniaturized data interface allows 8× parallel plate operation (768 total wells) within each incubator for enhanced throughputs. New electric field-based, multi-frequency measurement techniques capture >20 parameter images including tissue barrier, cell-surface attachment, cell flatness, and motility every 15 min throughout experiments. Using these real-time readouts, we characterized 16 cell types, ranging from primary epithelial to suspension, and quantified heterogeneity in mixed epithelial and mesenchymal co-cultures. A proof-of-concept screen of 904 diverse compounds using 13 semiconductor microplates demonstrates the platform's capability for mechanism of action (MOA) profiling with 25 distinct responses identified. The scalability of the semiconductor platform combined with the translatability of the high dimensional live-cell functional parameters expands high-throughput MOA profiling and phenotypic drug discovery applications.
Collapse
|
12
|
Prioritization of Microorganisms Isolated from the Indian Ocean Sponge Scopalina hapalia Based on Metabolomic Diversity and Biological Activity for the Discovery of Natural Products. Microorganisms 2023; 11:microorganisms11030697. [PMID: 36985270 PMCID: PMC10057949 DOI: 10.3390/microorganisms11030697] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 02/27/2023] [Accepted: 03/01/2023] [Indexed: 03/11/2023] Open
Abstract
Despite considerable advances in medicine and technology, humanity still faces many deadly diseases such as cancer and malaria. In order to find appropriate treatments, the discovery of new bioactive substances is essential. Therefore, research is now turning to less frequently explored habitats with exceptional biodiversity such as the marine environment. Many studies have demonstrated the therapeutic potential of bioactive compounds from marine macro- and microorganisms. In this study, nine microbial strains isolated from an Indian Ocean sponge, Scopalina hapalia, were screened for their chemical potential. The isolates belong to different phyla, some of which are already known for their production of secondary metabolites, such as the actinobacteria. This article aims at describing the selection method used to identify the most promising microorganisms in the field of active metabolites production. The method is based on the combination of their biological and chemical screening, coupled with the use of bioinformatic tools. The dereplication of microbial extracts and the creation of a molecular network revealed the presence of known bioactive molecules such as staurosporin, erythromycin and chaetoglobosins. Molecular network exploration indicated the possible presence of novel compounds in clusters of interest. The biological activities targeted in the study were cytotoxicity against the HCT-116 and MDA-MB-231 cell lines and antiplasmodial activity against Plasmodium falciparum 3D7. Chaetomium globosum SH-123 and Salinispora arenicola SH-78 strains actually showed remarkable cytotoxic and antiplasmodial activities, while Micromonospora fluostatini SH-82 demonstrated promising antiplasmodial effects. The ranking of the microorganisms as a result of the different screening steps allowed the selection of a promising strain, Micromonospora fluostatini SH-82, as a premium candidate for the discovery of new drugs.
Collapse
|
13
|
Garcia JA, Chen R, Xu M, Comerford SA, Hammer RE, Melton SD, Feagins LA. Acss2/HIF-2 signaling facilitates colon cancer growth and metastasis. PLoS One 2023; 18:e0282223. [PMID: 36862715 PMCID: PMC9980813 DOI: 10.1371/journal.pone.0282223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 02/09/2023] [Indexed: 03/03/2023] Open
Abstract
The microenvironment of solid tumors is characterized by oxygen and glucose deprivation. Acss2/HIF-2 signaling coordinates essential genetic regulators including acetate-dependent acetyl CoA synthetase 2 (Acss2), Creb binding protein (Cbp), Sirtuin 1 (Sirt1), and Hypoxia Inducible Factor 2α (HIF-2α). We previously shown in mice that exogenous acetate augments growth and metastasis of flank tumors derived from fibrosarcoma-derived HT1080 cells in an Acss2/HIF-2 dependent manner. Colonic epithelial cells are exposed to the highest acetate levels in the body. We reasoned that colon cancer cells, like fibrosarcoma cells, may respond to acetate in a pro-growth manner. In this study, we examine the role of Acss2/HIF-2 signaling in colon cancer. We find that Acss2/HIF-2 signaling is activated by oxygen or glucose deprivation in two human colon cancer-derived cell lines, HCT116 and HT29, and is crucial for colony formation, migration, and invasion in cell culture studies. Flank tumors derived from HCT116 and HT29 cells exhibit augmented growth in mice when supplemented with exogenous acetate in an Acss2/HIF-2 dependent manner. Finally, Acss2 in human colon cancer samples is most frequently localized in the nucleus, consistent with it having a signaling role. Targeted inhibition of Acss2/HIF-2 signaling may have synergistic effects for some colon cancer patients.
Collapse
Affiliation(s)
- Joseph A. Garcia
- Department of Medicine, Columbia University Medical Center, New York, New York, United States of America
- Research & Development, James J. Peters Veterans Affairs Medical Center, New York, New York, United States of America
| | - Rui Chen
- Department of Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Min Xu
- Department of Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Sarah A. Comerford
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Robert E. Hammer
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Shelby D. Melton
- Pathology & Laboratory Medicine, Veterans Affairs North Texas Health Care System, Dallas, Texas, United States of America
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Linda A. Feagins
- Department of Internal Medicine, Dell Medical School, The University of Texas at Austin, Austin, Texas, United States of America
| |
Collapse
|
14
|
Rahman Y, Afrin S, Perwez A, Ansari MO, Sarwar T, Ahmed S, Rizvi MA, Shadab GGHA, Tabish M. Nizatidine interacts with ct-DNA causing genotoxicity and cytotoxicity: an assessment by in vitro, in vivo, and in silico studies. J Biomol Struct Dyn 2023; 41:538-549. [PMID: 34856883 DOI: 10.1080/07391102.2021.2008496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
H2 receptor antagonists are the medication given for treating stomach ulcers, but lately, reports have shown their role in healing several malignant ulcers. The present work entails the interaction of H2 blocker nizatidine with calf thymus (ct)-DNA for determining the binding mode and energetics of the interaction. Multi-spectroscopic, calorimetric, viscometric and bioinformatic analysis revealed that nizatidine interacted with ct-DNA via groove-binding mode and is characterised by exothermic reaction. Moreover, assessment of genotoxic potential of nizatidine in vitro was carried out in peripheral human lymphocytes by alkaline comet assay. DNA damage occurred at high concentrations of nizatidine. Genotoxicity of nizatidine was also evaluated in vivo by assessing cytogenetic biomarkers viz. micronuclei formation and chromosomal aberration test. Nizatidine was able to induce micronuclei formation and chromosomal damage at high dose. Additionally, cytotoxic activity of nizatidine was determined in cancer cell lines, namely HeLa and HCT-116 and compared with the normal human cell line HEK-293 employing MTT assay. It was observed that nizatidine was more toxic towards HeLa and HCT-116 than HEK-293. Cell morphology analysis by compound inverted microscopy further strengthens the finding obtained through MTT assay.
Collapse
Affiliation(s)
- Yusra Rahman
- Department of Biochemistry, Faculty of Life Sciences, A.M. University, Aligarh, India
| | - Shumaila Afrin
- Department of Biochemistry, Faculty of Life Sciences, A.M. University, Aligarh, India
| | - Ahmad Perwez
- Department of Biosciences, Jamia Millia Islamia, New Delhi
| | - Mohd Owais Ansari
- Department of Zoology, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Tarique Sarwar
- Department of Biosciences, Jamia Millia Islamia, New Delhi
| | - Shahbaz Ahmed
- Department of Biochemistry, Faculty of Life Sciences, A.M. University, Aligarh, India
| | | | - G G Hammad A Shadab
- Department of Zoology, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Mohammad Tabish
- Department of Biochemistry, Faculty of Life Sciences, A.M. University, Aligarh, India
| |
Collapse
|
15
|
A zebrafish HCT116 xenograft model to predict anandamide outcomes on colorectal cancer. Cell Death Dis 2022; 13:1069. [PMID: 36564370 PMCID: PMC9789132 DOI: 10.1038/s41419-022-05523-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 12/08/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022]
Abstract
Colon cancer is one of the leading causes of death worldwide. In recent years, cannabinoids have been extensively studied for their potential anticancer effects and symptom management. Several in vitro studies reported anandamide's (AEA) ability to block cancer cell proliferation and migration, but evidence from in vivo studies is still lacking. Thus, in this study, the effects of AEA exposure in zebrafish embryos transplanted with HCT116 cells were evaluated. Totally, 48 hpf xenografts were exposed to 10 nM AEA, 10 nM AM251, one of the cannabinoid 1 receptor (CB1) antagonist/inverse agonists, and to AEA + AM251, to verify the specific effect of AEA treatment. AEA efficacy was evaluated by confocal microscopy, which demonstrated that these xenografts presented a smaller tumor size, reduced tumor angiogenesis, and lacked micrometastasis formation. To gain deeper evidence into AEA action, microscopic observations were completed by molecular analyses. RNA seq performed on zebrafish transcriptome reported the downregulation of genes involved in cell proliferation, angiogenesis, and the immune system. Conversely, HCT116 cell transcripts resulted not affected by AEA treatment. In vitro HCT116 culture, in fact, confirmed that AEA exposure did not affect cell proliferation and viability, thus suggesting that the reduced tumor size mainly depends on direct effects on the fish rather than on the transplanted cancer cells. AEA reduced cell proliferation and tumor angiogenesis, as suggested by socs3 and pcnp mRNAs and Vegfc protein levels, and exerted anti-inflammatory activity, as indicated by the reduction of il-11a, mhc1uba, and csf3b mRNA. Of note, are the results obtained in groups exposed to AM251, which presence nullifies AEA's beneficial effects. In conclusion, this study promotes the efficacy of AEA in personalized cancer therapy, as suggested by its ability to drive tumor growth and metastasis, and strongly supports the use of zebrafish xenograft as an emerging model platform for cancer studies.
Collapse
|
16
|
Utharala R, Grab A, Vafaizadeh V, Peschke N, Ballinger M, Turei D, Tuechler N, Ma W, Ivanova O, Ortiz AG, Saez-Rodriguez J, Merten CA. A microfluidic Braille valve platform for on-demand production, combinatorial screening and sorting of chemically distinct droplets. Nat Protoc 2022; 17:2920-2965. [PMID: 36261631 DOI: 10.1038/s41596-022-00740-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 06/16/2022] [Indexed: 11/09/2022]
Abstract
Droplet microfluidics is a powerful tool for a variety of biological applications including single-cell genetics, antibody discovery and directed evolution. All these applications make use of genetic libraries, illustrating the difficulty of generating chemically distinct droplets for screening applications. This protocol describes our Braille Display valving platform for on-demand generation of droplets with different chemical contents (16 different reagents and combinations thereof), as well as sorting droplets with different chemical properties, on the basis of fluorescence signals. The Braille Display platform is compact, versatile and cost efficient (only ~US$1,000 on top of a standard droplet microfluidics setup). The procedure includes manufacturing of microfluidic chips, assembly of custom hardware, co-encapsulation of cells and drugs into droplets, fluorescence detection of readout signals and data analysis using shared, freely available LabVIEW and Python packages. As a first application, we demonstrate the complete workflow for screening cancer cell drug sensitivities toward 74 conditions. Furthermore, we describe here an assay enabling the normalization of the observed drug sensitivity to the number of cancer cells per droplet, which additionally increases the robustness of the system. As a second application, we also demonstrate the sorting of droplets according to enzymatic activity. The drug screening application can be completed within 2 d; droplet sorting takes ~1 d; and all preparatory steps for manufacturing molds, chips and setting up the Braille controller can be accomplished within 1 week.
Collapse
Affiliation(s)
- Ramesh Utharala
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Anna Grab
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- Clinical Cooperation Unit Molecular Hematology/Oncology, DKFZ Heidelberg and Translational Myeloma Research Group, Department of Internal Medicine V, University Hospital, Heidelberg, Germany
| | - Vida Vafaizadeh
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- Institute of Bioengineering, School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Nicolas Peschke
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Martine Ballinger
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- Collaboration for joint PhD degree between EMBL and Heidelberg University, Faculty of Biosciences, Heidelberg, Germany
| | - Denes Turei
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- Faculty of Medicine and Heidelberg University Hospital, Institute of Computational Biomedicine, Heidelberg University, Heidelberg, Germany
| | - Nadine Tuechler
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Wenwei Ma
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Olga Ivanova
- Faculty of Medicine and Heidelberg University Hospital, Institute of Computational Biomedicine, Heidelberg University, Heidelberg, Germany
| | | | - Julio Saez-Rodriguez
- Faculty of Medicine and Heidelberg University Hospital, Institute of Computational Biomedicine, Heidelberg University, Heidelberg, Germany
- Faculty of Medicine, Joint Research Centre for Computational Biomedicine (JRC-COMBINE), RWTH Aachen University, Aachen, Germany
| | - Christoph A Merten
- Institute of Bioengineering, School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| |
Collapse
|
17
|
Saggese A, Giglio R, D’Anzi N, Baccigalupi L, Ricca E. Comparative Genomics and Physiological Characterization of Two Aerobic Spore Formers Isolated from Human Ileal Samples. Int J Mol Sci 2022; 23:14946. [PMID: 36499272 PMCID: PMC9739757 DOI: 10.3390/ijms232314946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/25/2022] [Accepted: 11/25/2022] [Indexed: 12/05/2022] Open
Abstract
Spore formers are ubiquitous microorganisms commonly isolated from most environments, including the gastro-intestinal tract (GIT) of insects and animals. Spores ingested as food and water contaminants safely transit the stomach and reach the intestine, where some of them germinate and temporarily colonize that niche. In the lower part of the GIT, they re-sporulate and leave the body as spores, therefore passing through their entire life cycle in the animal body. In the intestine, both un-germinated spores and germination-derived cells interact with intestinal and immune cells and have health-beneficial effects, which include the production of useful compounds, protection against pathogenic microorganisms, contribution to the development of an efficient immune system and modulation of the gut microbial composition. We report a genomic and physiological characterization of SF106 and SF174, two aerobic spore former strains previously isolated from ileal biopsies of healthy human volunteers. SF106 and SF174 belong respectively to the B. subtilis and Alkalihalobacillus clausii (formerly Bacillus clausii) species, are unable to produce toxins or other metabolites with cytotoxic activity against cultured human cells, efficiently bind mucin and human epithelial cells in vitro and produce molecules with antimicrobial and antibiofilm activities.
Collapse
Affiliation(s)
- Anella Saggese
- Department of Biology, Federico II University of Naples, 80125 Naples, Italy
| | | | | | - Loredana Baccigalupi
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, 80131 Naples, Italy
| | - Ezio Ricca
- Department of Biology, Federico II University of Naples, 80125 Naples, Italy
| |
Collapse
|
18
|
Tolstik E, Gongalsky MB, Dierks J, Brand T, Pernecker M, Pervushin NV, Maksutova DE, Gonchar KA, Samsonova JV, Kopeina G, Sivakov V, Osminkina LA, Lorenz K. Raman and fluorescence micro-spectroscopy applied for the monitoring of sunitinib-loaded porous silicon nanocontainers in cardiac cells. Front Pharmacol 2022; 13:962763. [PMID: 36016563 PMCID: PMC9397571 DOI: 10.3389/fphar.2022.962763] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 07/05/2022] [Indexed: 11/17/2022] Open
Abstract
Nanomaterials are a central pillar in modern medicine. They are thought to optimize drug delivery, enhance therapeutic efficacy, and reduce side-effects. To foster this technology, analytical methods are needed to validate not only the localization and distribution of these nanomaterials, but also their compatibility with cells, drugs, and drug release. In the present work, we assessed nanoparticles based on porous silicon (pSiNPs) loaded with the clinically used tyrosine kinase inhibitor sunitinib for their effectiveness of drug delivery, release, and toxicity in colon cancer cells (HCT 116 cells) and cardiac myoblast cells (H9c2) using Raman micro-spectroscopy, high-resolution fluorescence microscopy, along with biological methods for toxicological effects. We produced pSiNPs with a size of about 100 nm by grinding mesoporous silicon layers. pSiNPs allowed an effective loading of sunitinib due to their high porosity. Photoluminescence properties of the nanoparticles within the visible spectrum allowed the visualization of their uptake in cardiac cells. Raman micro-spectroscopy allowed not only the detection of the uptake and distribution of pSiNPs within the cells via a characteristic silicon Raman band at about 518–520 cm−1, but also the localization of the drug based on its characteristic molecular fingerprints. Cytotoxicity studies by Western blot analyses of apoptotic marker proteins such as caspase-3, and the detection of apoptosis by subG1-positive cell fractions in HCT 116 and MTT analyses in H9c2 cells, suggest a sustained release of sunitinib from pSiNPs and delayed cytotoxicity of sunitinib in HCT 116 cells. The analyses in cardiac cells revealed that pSiNPs are well tolerated and that they may even protect from toxic effects in these cells to some extent. Analyses of the integrity of mitochondrial networks as an early indicator for apoptotic cellular effects seem to validate these observations. Our study suggests pSiNPs-based nanocontainers for efficient and safe drug delivery and Raman micro-spectroscopy as a reliable method for their detection and monitoring. Thus, the herein presented nanocontainers and analytical methods have the potential to allow an efficient advancement of nanoparticles for targeted and sustained intracellular drug release that is of need, e.g., in chronic diseases and for the prevention of cardiac toxicity.
Collapse
Affiliation(s)
- E. Tolstik
- Leibniz-Institut für Analytische Wissenschaften—ISAS—e.V., Dortmund, Germany
- *Correspondence: E. Tolstik, elen.tolstik@isas; L. A. Osminkina, ; K. Lorenz,
| | - M. B. Gongalsky
- Lomonosov Moscow State University, Faculty of Physics, Moscow, Russia
| | - J. Dierks
- Leibniz-Institut für Analytische Wissenschaften—ISAS—e.V., Dortmund, Germany
| | - T. Brand
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
| | - M. Pernecker
- Leibniz-Institut für Analytische Wissenschaften—ISAS—e.V., Dortmund, Germany
| | - N. V. Pervushin
- Lomonosov Moscow State University, Faculty of Medicine, Moscow, Russia
| | - D. E. Maksutova
- Lomonosov Moscow State University, Faculty of Physics, Moscow, Russia
| | - K. A. Gonchar
- Lomonosov Moscow State University, Faculty of Physics, Moscow, Russia
| | - J. V. Samsonova
- Lomonosov Moscow State University, Faculty of Chemistry, Moscow, Russia
| | - G. Kopeina
- Lomonosov Moscow State University, Faculty of Medicine, Moscow, Russia
| | - V. Sivakov
- Leibniz Institute of Photonic Technology, Department Functional Interfaces, Jena, Germany
| | - L. A. Osminkina
- Lomonosov Moscow State University, Faculty of Physics, Moscow, Russia
- Institute for Biological Instrumentation of Russian Academy of Sciences, Moscow, Russia
- *Correspondence: E. Tolstik, elen.tolstik@isas; L. A. Osminkina, ; K. Lorenz,
| | - K. Lorenz
- Leibniz-Institut für Analytische Wissenschaften—ISAS—e.V., Dortmund, Germany
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
- Comprehensive Heart Failure Center, University Hospital of Würzburg, Würzburg, Germany
- *Correspondence: E. Tolstik, elen.tolstik@isas; L. A. Osminkina, ; K. Lorenz,
| |
Collapse
|
19
|
Study on the Action Mechanism of the Yifei Jianpi Tongfu Formula in Treatment of Colorectal Cancer Lung Metastasis Based on Network Analysis, Molecular Docking, and Experimental Validation. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:6229444. [PMID: 35942366 PMCID: PMC9356795 DOI: 10.1155/2022/6229444] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 06/28/2022] [Indexed: 11/18/2022]
Abstract
Objective The lung is the second most common site of colorectal cancer (CRC) metastasis. This study aims to investigate the therapeutic effects and potential action mechanisms of Yifei Jianpi Tongfu formula (YJTF) in CRC lung metastasis in a comprehensive and systematic way by network analysis, molecular docking, and experimental verification. Methods The main ingredients in YJTF were screened from the Traditional Chinese Medicine System Pharmacology Database and Analysis Platform (TCMSP) and Traditional Chinese Medicine Integrated Database (TCMID), and the disease-related targets from the Online Mendelian Inheritance in Man (OMIM) and GeneCards and the compound-related targets from SwissTargetPrediction were collected. Then, Metascape was used for pathway annotation and enrichment analysis, and meanwhile, a protein-protein interaction (PPI) network was constructed. Molecular docking was carried out to investigate interactions between the active compounds and the potential targets. The in vivo effect of YJTF on CRC lung metastasis was observed in a tail vein injection mouse model. Results A total of 243 active compounds and 81 disease-related targets of YJTF were selected for analysis. The results of multiple network analysis showed that the core targets of YJTF were enriched onto various cancer-related pathways, especially focal adhesion and adherens junction. The results of molecular docking demonstrated that all core compounds (quercetin, kaempferol, luteolin, apigenin, and isorhamnetin) were capable of binding with AKT1, EGFR, SRC, ESR1, and PTGS2. Experimental validation in vivo demonstrated that YJTF combined with oxaliplatin could significantly reduce the number of lung metastases and improve the quality of life in mice. Further research suggested that YJTF inhibited CRC lung metastasis probably by modulating epithelial-to-mesenchymal transition (EMT). Conclusions According to the analysis, YJTF can be considered as an effective adjuvant therapy for CRC lung metastasis.
Collapse
|
20
|
Nairon KG, DePalma TJ, Zent JM, Leight JL, Skardal A. Tumor cell-conditioned media drives collagen remodeling via fibroblast and pericyte activation in an in vitro premetastatic niche model. iScience 2022; 25:104645. [PMID: 35811850 PMCID: PMC9257340 DOI: 10.1016/j.isci.2022.104645] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 05/25/2022] [Accepted: 06/15/2022] [Indexed: 11/21/2022] Open
Abstract
Primary tumors secrete large quantities of cytokines and exosomes into the bloodstream, which are uptaken at downstream sites and induce a pro-fibrotic, pro-inflammatory premetastatic niche. Niche development is associated with later increased metastatic burden, but the cellular and matrix changes in the niche that facilitate metastasis are yet unknown. Furthermore, there is no current standard model to study this phenomenon. Here, biofabricated collagen and hyaluronic acid hydrogel models were employed to identify matrix changes elicited by pericytes and fibroblasts after exposure to colorectal cancer-secreted factors. Focusing on myofibroblast activation and collagen remodeling, we report fibroblast activation and pericyte stunting in response to tumor signaling. In addition, we characterize contributions of both cell types to matrix dysregulation via collagen degradation, deposition, and architectural remodeling. With these findings, we discuss potential impacts on tissue stiffening and vascular leakiness and suggest pathways of interest for future mechanistic studies of metastatic cell-premetastatic niche interactions.
Collapse
Affiliation(s)
- Kylie G. Nairon
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Thomas J. DePalma
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Joshua M. Zent
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Jennifer L. Leight
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH 43210, USA
- The Ohio State University and Arthur G. James Comprehensive Cancer Center, Columbus, OH 43210, USA
- Center for Cancer Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Aleksander Skardal
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH 43210, USA
- The Ohio State University and Arthur G. James Comprehensive Cancer Center, Columbus, OH 43210, USA
- Center for Cancer Engineering, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
21
|
Potočnjak I, Šimić L, Vukelić I, Batičić L, Domitrović R. Oleanolic acid induces HCT116 colon cancer cell death through the p38/FOXO3a/Sirt6 pathway. Chem Biol Interact 2022; 363:110010. [PMID: 35690101 DOI: 10.1016/j.cbi.2022.110010] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 05/14/2022] [Accepted: 06/07/2022] [Indexed: 12/08/2022]
Abstract
Oleanolic acid (OA) is a natural compound that possesses numerous beneficial health effects, including anticancer activity. The current study aimed to investigate the role of forkhead box O3a (FOXO3a) in autophagy/mitophagy by OA in HCT116 cell line. OA dose-dependently reduced viability of HCT116 cells, with IC50 = 29.8 μΜ. The expression of cleaved caspase-3 and poly (ADP-ribose) polymerase 1 increased after OA treatment, suggesting induction of apoptosis. Concurrently, OA induced autophagy, evidenced by increased expression of Beclin-1, autophagy-related protein 5 and microtubule-associated protein1A/1B-light chain 3 beta (LC3B), which played a prosurvival role. The induction of mitophagy was suggested by increased expression of p62 and PTEN-induced kinase 1 and reduced expression of translocase of outer mitochondrial membrane 20, which colocalized with LC3B. OA also induced nuclear accumulation of forkhead box O3a (FOXO3a). The cytotoxic activity of OA coincided with upregulation of p38. Inhibition of p38 led to increase in FOXO3a and NAD+-dependent deacetylase sirtuin 6 expression. In vivo, OA inhibited tumor growth in colon cancer xenograft mice. Our results suggest concomitant induction of apoptosis and prosurvival mitophagy by OA in colon cancer via p38/FOXO3a/Sirt6 signaling. Additionally, our data demonstrate that OA can chemosensitize colon cancer cells to 5-fluorouracil (5-FU).
Collapse
Affiliation(s)
- Iva Potočnjak
- Department of Medical Chemistry, Biochemistry and Clinical Chemistry, Faculty of Medicine, University of Rijeka, Croatia
| | - Lidija Šimić
- Department of Medical Chemistry, Biochemistry and Clinical Chemistry, Faculty of Medicine, University of Rijeka, Croatia
| | - Iva Vukelić
- Department of Medical Chemistry, Biochemistry and Clinical Chemistry, Faculty of Medicine, University of Rijeka, Croatia
| | - Lara Batičić
- Department of Medical Chemistry, Biochemistry and Clinical Chemistry, Faculty of Medicine, University of Rijeka, Croatia
| | - Robert Domitrović
- Department of Medical Chemistry, Biochemistry and Clinical Chemistry, Faculty of Medicine, University of Rijeka, Croatia.
| |
Collapse
|
22
|
Chen YT, Tseng TT, Tsai HP, Huang MY. Arylquin 1 (Potent Par-4 Secretagogue) Inhibits Tumor Progression and Induces Apoptosis in Colon Cancer Cells. Int J Mol Sci 2022; 23:ijms23105645. [PMID: 35628455 PMCID: PMC9143413 DOI: 10.3390/ijms23105645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/14/2022] [Accepted: 05/17/2022] [Indexed: 11/16/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common gastrointestinal cancers worldwide. Current therapeutic strategies mainly involve surgery and chemoradiotherapy; however, novel antitumor compounds are needed to avoid drug resistance in CRC, as well as the severe side effects of current treatments. In this study, we investigated the anticancer effects and underlying mechanisms of Arylquin 1 in CRC. The MTT assay was used to detect the viability of SW620 and HCT116 cancer cells treated with Arylquin 1 in a dose-dependent manner in vitro. Further, wound-healing and transwell migration assays were used to evaluate the migration and invasion abilities of cultured cells, and Annexin V was used to detect apoptotic cells. Additionally, Western blot was used to identify the expression levels of N-cadherin, caspase-3, cyclin D1, p-extracellular signal-regulated kinase (ERK), p-c-JUN N-terminal kinase (JNK), and phospho-p38, related to key signaling proteins, after administration of Arylquin 1. Xenograft experiments further confirmed the effects of Arylquin 1 on CRC cells in vivo. Arylquin 1 exhibited a dose-dependent reduction in cell viability in cultured CRC cells. It also inhibited cell proliferation, migration, and invasion, and induced apoptosis. Mechanistic analysis demonstrated that Arylquin 1 increased phosphorylation levels of ERK, JNK, and p38. In a mouse xenograft model, Arylquin 1 treatment diminished the growth of colon tumors after injection of cultured cancer cells. Arylquin 1 may have potential anticancer effects and translational significance in the treatment of CRC.
Collapse
Affiliation(s)
- Yi-Ting Chen
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan; (Y.-T.C.); (T.-T.T.)
- Department of Pathology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Tzu-Ting Tseng
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan; (Y.-T.C.); (T.-T.T.)
| | - Hung-Pei Tsai
- Department of Surgery, Division of Neurosurgery, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan;
| | - Ming-Yii Huang
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- Department of Radiation Oncology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Correspondence:
| |
Collapse
|
23
|
Kajsik M, Chovancova B, Liskova V, Babula P, Krizanova O. Slow sulfide donor GYY4137 potentiates effect of paclitaxel on colorectal carcinoma cells. Eur J Pharmacol 2022; 922:174875. [DOI: 10.1016/j.ejphar.2022.174875] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 02/15/2022] [Accepted: 03/07/2022] [Indexed: 11/03/2022]
|
24
|
Liskova V, Kajsik M, Chovancova B, Roller L, Krizanova O. Camptothecin, triptolide, and apoptosis inducer kit have differential effects on mitochondria in colorectal carcinoma cells. FEBS Open Bio 2022; 12:913-924. [PMID: 35318813 PMCID: PMC9063445 DOI: 10.1002/2211-5463.13401] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/16/2021] [Accepted: 03/21/2022] [Indexed: 11/17/2022] Open
Abstract
Mitochondrial fission and fusion are required for cell survival, and several studies have shown an imbalance between fission and fusion in cancer. High levels of mitochondrial fusion are observed in drug‐resistant tumor cells, whereas mitochondrial fission may be important in sensitizing tumor cells to chemotherapy drugs. Based on current knowledge, we hypothesized that different chemotherapeutics might differentially affect mitochondrial dynamics and energy production. Thus, we selected chemotherapeutics with different mechanisms of action (camptothecin, triptolide and apoptosis inducer kit) and investigated their effect on mitochondria in colorectal carcinoma cells. We report that these chemotherapeutics decreased the activity of complex I and reduced the mitochondrial membrane potential, and also decreased the size of mitochondria in the colorectal carcinoma cell lines DLD1 and HCT‐116. Treatment with camptothecin, triptolide and/or apoptosis inducer kit results in differential effects of fission on apoptosis in these cells. Our results suggest that fission is an important process in apoptosis induced by chemotherapeutics.
Collapse
Affiliation(s)
- Veronika Liskova
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Marek Kajsik
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia.,Department of Chemistry, Faculty of Natural Sciences, University of Ss. Cyril and Methodius, Trnava, Slovakia
| | - Barbora Chovancova
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Ladislav Roller
- Institute of Zoology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Olga Krizanova
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia.,Department of Chemistry, Faculty of Natural Sciences, University of Ss. Cyril and Methodius, Trnava, Slovakia
| |
Collapse
|
25
|
Corona SP, Walker F, Weinstock J, Lessene G, Faux M, Burgess AW. Dual drug targeting to kill colon cancers. Cancer Med 2022; 11:2612-2626. [PMID: 35301819 PMCID: PMC9249985 DOI: 10.1002/cam4.4641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 01/06/2022] [Accepted: 02/09/2022] [Indexed: 11/18/2022] Open
Abstract
Introduction Colorectal cancer (CRC) is driven by a small set of oncogenic and tumour suppressor mutations. However, different combinations of mutations often lead to poor tumour responses to individual anticancer drugs. We have investigated the antiproliferative and in vitro cytotoxic activity of pair‐wise combinations of inhibitors which target specific signalling pathways in colon cancer cells. Objectives To target specific signaling pathways pairwise with inhibitors in order to kill colon cancer cells. Methods The effects of different concentrations of two inhibitors on the proliferation and viability of colon cancer cell lines were measured using cell titre glow and cytotoxic assays in 2D and 3D cell micro‐cultures. One successful drug combination was used to treat a colon cancer cell line growing as a xenograft in nude mice. Results Colon cancer cells in non‐adherent cultures were killed more effectively by combinations of pyrvinium pamoate (a Wnt pathway inhibitor) and ABT263 (a pro‐apoptotic Bcl‐2 family inhibitor) or Ly29004 (a PI3kinase inhibitor). However, in a mouse xenograft model, the formulation and toxicity of the ABT737/PP combination prevent the use of these drugs for treatment of tumours. Fortunately, oral analogues of PP (pyrvinium phosphate, PPh) and ABT737(ABT263) have equivalent activity and can be used for treatment of mice carrying SW620 colorectal cancer xenografts. The PPh/ABT263 induced SW620 tumour cell apoptosis and reduced the rate of SW620 tumour growth. Conclusion By combining a Wnt signaling inhibitor (pyrvinium phosphate) and a pro‐survival inhibitor (ABT263) colon cancer cells can be killed. Combinations of Wnt signalling inhibitors with an inhibitor of the Bcl pro‐survival protein family should be considered for the treatment of patients with precancerous colon adenomas or advanced colorectal cancers with APC mutations.
Collapse
Affiliation(s)
- Silvia Paola Corona
- Structural Biology Division, WEHI, Parkville, Australia.,Personalised Oncology Division, WEHI, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia.,Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, Australia.,Ludwig Institute for Cancer Research, Parkville, Australia
| | - Francesca Walker
- Structural Biology Division, WEHI, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia.,Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, Australia.,Ludwig Institute for Cancer Research, Parkville, Australia
| | - Janet Weinstock
- Structural Biology Division, WEHI, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia.,Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, Australia.,Ludwig Institute for Cancer Research, Parkville, Australia
| | - Guillaume Lessene
- Department of Medical Biology, University of Melbourne, Parkville, Australia.,Chemical Biology Division, WEHI, Parkville, Australia.,Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, Australia
| | - Maree Faux
- Structural Biology Division, WEHI, Parkville, Australia.,Personalised Oncology Division, WEHI, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia.,Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, Australia.,Ludwig Institute for Cancer Research, Parkville, Australia.,Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Antony W Burgess
- Structural Biology Division, WEHI, Parkville, Australia.,Personalised Oncology Division, WEHI, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia.,Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, Australia.,Ludwig Institute for Cancer Research, Parkville, Australia
| |
Collapse
|
26
|
Han SP, Scherer L, Gethers M, Salvador AM, Salah MBH, Mancusi R, Sagar S, Hu R, DeRogatis J, Kuo YH, Marcucci G, Das S, Rossi JJ, Goddard WA. Programmable siRNA pro-drugs that activate RNAi activity in response to specific cellular RNA biomarkers. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 27:797-809. [PMID: 35116191 PMCID: PMC8789579 DOI: 10.1016/j.omtn.2021.12.039] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 12/31/2021] [Indexed: 11/13/2022]
Abstract
Since Paul Ehrlich's introduction of the "magic bullet" concept in 1908, drug developers have been seeking new ways to target drug activity to diseased cells while limiting effects on normal tissues. In recent years, it has been proposed that coupling riboswitches capable of detecting RNA biomarkers to small interfering RNAs (siRNAs) to create siRNA pro-drugs could selectively activate RNA interference (RNAi) activity in specific cells. However, this concept has not been achieved previously. We report here that we have accomplished this goal, validating a simple and programmable new design that functions reliably in mammalian cells. We show that these conditionally activated siRNAs (Cond-siRNAs) can switch RNAi activity against different targets between clearly distinguished OFF and ON states in response to different cellular RNA biomarkers. Notably, in a rat cardiomyocyte cell line (H9C2), one version of our construct demonstrated biologically meaningful inhibition of a heart-disease-related target gene protein phosphatase 3 catalytic subunit alpha (PPP3CA) in response to increased expression of the pathological marker atrial natriuretic peptide (NPPA) messenger RNA (mRNA). Our results demonstrate the ability of synthetic riboswitches to regulate gene expression in mammalian cells, opening a new path for development of programmable siRNA pro-drugs.
Collapse
Affiliation(s)
- Si-ping Han
- Materials and Process Simulation Center, California Institute of Technology, Pasadena, CA 91125, USA
- Department of Molecular and Cellular Biology, City of Hope, Duarte, CA 91010, USA
| | - Lisa Scherer
- Department of Molecular and Cellular Biology, City of Hope, Duarte, CA 91010, USA
| | - Matt Gethers
- Materials and Process Simulation Center, California Institute of Technology, Pasadena, CA 91125, USA
| | - Ane M. Salvador
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Marwa Ben Haj Salah
- Department of Molecular and Cellular Biology, City of Hope, Duarte, CA 91010, USA
| | - Rebecca Mancusi
- Department of Molecular and Cellular Biology, City of Hope, Duarte, CA 91010, USA
| | - Sahil Sagar
- Department of Molecular and Cellular Biology, City of Hope, Duarte, CA 91010, USA
| | - Robin Hu
- Department of Molecular and Cellular Biology, City of Hope, Duarte, CA 91010, USA
| | - Julia DeRogatis
- Department of Molecular and Cellular Biology, City of Hope, Duarte, CA 91010, USA
| | - Ya-Huei Kuo
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA 91010, USA
| | - Guido Marcucci
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA 91010, USA
| | - Saumya Das
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | - John J. Rossi
- Department of Molecular and Cellular Biology, City of Hope, Duarte, CA 91010, USA
| | - William A. Goddard
- Materials and Process Simulation Center, California Institute of Technology, Pasadena, CA 91125, USA
| |
Collapse
|
27
|
(E)-3-Arylidene-4-diazopyrrolidine-2,5-diones conveniently elaborated into cytotoxic compounds bearing primary sulfonamide and Michael acceptor moieties. MENDELEEV COMMUNICATIONS 2022. [DOI: 10.1016/j.mencom.2022.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
28
|
Yuan C, Zhao X, Wangmo D, Alshareef D, Gates TJ, Subramanian S. Tumor models to assess immune response and tumor-microbiome interactions in colorectal cancer. Pharmacol Ther 2022; 231:107981. [PMID: 34480964 PMCID: PMC8844062 DOI: 10.1016/j.pharmthera.2021.107981] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 08/20/2021] [Accepted: 08/24/2021] [Indexed: 02/07/2023]
Abstract
Despite significant advances over the past 2 decades in preventive screening and therapy aimed at improving patient survival, colorectal cancer (CRC) remains the second most common cause of cancer death in the United States. The average 5-year survival rate of CRC patients with positive regional lymph nodes is only 40%, while less than 5% of patients with distant metastases survive beyond 5 years. There is a critical need to develop novel therapies that can improve overall survival in patients with poor prognoses, particularly since 60% of them are diagnosed at an advanced stage. Pertinently, immune checkpoint blockade therapy has dramatically changed how we treat CRC patients with microsatellite-instable high tumors. Furthermore, accumulating evidence shows that changes in gut microbiota are associated with the regulation of host antitumor immune response and cancer progression. Appropriate animal models are essential to deciphering the complex mechanisms of host antitumor immune response and tumor-gut microbiome metabolic interactions. Here, we discuss various mouse models of colorectal cancer that are developed to address key questions on tumor immune response and tumor-microbiota interactions. These CRC models will also serve as resourceful tools for effective preclinical studies.
Collapse
Affiliation(s)
- Ce Yuan
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Xianda Zhao
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Dechen Wangmo
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455, United States of America; Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Duha Alshareef
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Travis J Gates
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455, United States of America; Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Subbaya Subramanian
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455, United States of America; Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, United States of America; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, United States of America.
| |
Collapse
|
29
|
Martínez-Iglesias O, Carrera I, Naidoo V, Cacabelos R. AntiGan: An Epinutraceutical Bioproduct with Antitumor Properties in Cultured Cell Lines. Life (Basel) 2022; 12:97. [PMID: 35054489 PMCID: PMC8780983 DOI: 10.3390/life12010097] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/16/2021] [Accepted: 01/01/2022] [Indexed: 12/13/2022] Open
Abstract
Novel and effective chemotherapeutic agents are needed to improve cancer treatment. Epidrugs are currently used for cancer therapy but also exhibit toxicity. Targeting the epigenetic apparatus with bioproducts may aid cancer prevention and treatment. To determine whether the lipoprotein marine extract AntiGan shows epigenetic and antitumor effects, cultured HepG2 (hepatocellular carcinoma) and HCT116 (colorectal carcinoma) cell lines were treated with AntiGan (10, 50, 100, and to 500 µg/mL) for 24 h, 48 h, and 72 h. AntiGan (10 µg/mL) reduced cell viability after 48 h and increased Bax expression; AntiGan (10 and 50 µg/mL) increased caspase-3 immunoreactivity in HepG2 and HCT116 cells. AntiGan (10 and 50 µg/mL) attenuated COX-2 and IL-17 expression in both cell lines. AntiGan (10 µg/mL) increased 5mC levels in both cell types and reduced DNMT1 and DNMT3a expression in these cells. AntiGan (10 and 50 µg/mL) promoted DNMT3a immunoreactivity and reduced SIRT1 mRNA expression in both cell types. In HCT116 cells treated with AntiGan (10 µg/mL), SIRT1 immunoreactivity localized to nuclei and the cytoplasm; AntiGan (50 µg/mL) increased cytoplasmic SIRT1 localization in HCT116 cells. AntiGan is a novel antitumoral bioproduct with epigenetic properties (epinutraceutical) for treating liver and colorectal cancer.
Collapse
Affiliation(s)
- Olaia Martínez-Iglesias
- EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine, 15165 Bergondo, Corunna, Spain; (I.C.); (V.N.); (R.C.)
| | | | | | | |
Collapse
|
30
|
Bredeck G, Halamoda-Kenzaoui B, Bogni A, Lipsa D, Bremer-Hoffmann S. Tiered testing of micro- and nanoplastics using intestinal in vitro models to support hazard assessments. ENVIRONMENT INTERNATIONAL 2022; 158:106921. [PMID: 34634620 DOI: 10.1016/j.envint.2021.106921] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/01/2021] [Accepted: 10/04/2021] [Indexed: 06/13/2023]
Abstract
The uncertainty of potential risks associated with micro- and nanoplastics (MNPs) are of growing public concern. However, the diversity of MNPs in the environment makes a systematic analysis of potential health effects challenging. New tools and approaches are necessary to investigate biological effects of MNPs. With this quick scoping review, we aim to analyse the suitability of in vitro models for assessing the interaction of MNPs with intestinal cells. Our analysis revealed that currently the majority of in vitro tests are based on the three cell lines Caco-2, HT-29, and HCT-116. They have particularly been used to assess endpoints related to basal cytotoxicity, the internalisation of MNPs and effects on the intestinal barrier. When co-cultured with various cell lines, they also allow to investigate additional effects such as inflammation, metabolic actions and the relevance of the intestinal mucus. However, methodological gaps remain regarding the assessment of a potential accumulation of MNPs, leaching of additives/impurities and in resulting long-term effects as well as cell-type specific toxicities. In addition, only few in vitro studies investigated effects of MNPs on the microbiome. Stem cell-based assays using, for example, the emerging organoid technology are promising for analysing MNP effects on tissue-like structures, while avoiding the particular characteristics of the currently used cancer derived cell lines. The various cell lines and culture techniques can be combined in testing strategies, to better elucidate potential biological interaction of MNPs with biological systems. We suggest to implement a tiered testing strategy, in which monocultures can serve as a tool for high-throughput testing of MNPs. In the next steps co-cultures can be used to assess the potential of a systemic uptake of MNPs and organ-on-a-chip models will provide more reliable insights into relevant doses triggering biological effects. Finally, organoids can help to discover new and more complex reactions initiated by MNPs.
Collapse
Affiliation(s)
- Gerrit Bredeck
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | | | - Alessia Bogni
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Dorelia Lipsa
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | | |
Collapse
|
31
|
Abstract
Genetically engineered Salmonella typhimurium can specifically colonize tumor tissues and drastically inhibit tumor growth. Vibrio vulnificus flagellin B (FlaB), a natural ligand of Toll-like receptor 5 (TLR5) that can activate robust host immune system, is an excellent adjuvant for cancer immunotherapy with high binding affinity to TLR5. Here, we constructed attenuated S. typhimurium that expresses flagellin B (FlaB) with a controlled expression system to enhance targeted cancer immunotherapy with increased good safety profiles. Visualized therapy can also be achieved with bioluminescence imaging by introducing the lux operon into the attenuated Salmonella.
Collapse
Affiliation(s)
- Yujie Sun
- School of Biomedical Sciences, Hunan University, Changsha, China
| | - Jin Hai Zheng
- School of Biomedical Sciences, Hunan University, Changsha, China.
| |
Collapse
|
32
|
Hajdowska K, Student S, Borys D. Graph based method for cell segmentation and detection in live-cell fluorescence microscope imaging. Biomed Signal Process Control 2022. [DOI: 10.1016/j.bspc.2021.103071] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
33
|
Leonard NA, Reidy E, Thompson K, McDermott E, Peerani E, Tomas Bort E, Balkwill FR, Loessner D, Ryan AE. Stromal Cells Promote Matrix Deposition, Remodelling and an Immunosuppressive Tumour Microenvironment in a 3D Model of Colon Cancer. Cancers (Basel) 2021; 13:cancers13235998. [PMID: 34885111 PMCID: PMC8656544 DOI: 10.3390/cancers13235998] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/24/2021] [Accepted: 11/25/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Colorectal cancer is the third most common type of cancer in the world. Immune cells and normal supporting cells (MSCs) within a tumour affect patient survival and change how well treatments work. This research aimed to develop a more relevant 3D cancer model that combines MSCs and immune cells with cancer cells to test the effects of multiple cell types on tumour growth. We successfully developed a 3D model that shows that MSCs and immune cells can change the cancer-supporting environment around the tumour cells. We show that combining MSCs and immune cells with cancer cells can increase the level of immune-suppressing molecules they release and change immunotherapeutic drug targets on the cancer cells, similar to changes seen in human tumours. Using this 3D model for research may be better for testing new drugs than traditional 2D methods and could enable the identification of new drug targets. Abstract Colorectal cancer (CRC) is the third leading cause of cancer-related deaths worldwide. CRC develops in a complex tumour microenvironment (TME) with both mesenchymal stromal cells (MSCs) and immune infiltrate, shown to alter disease progression and treatment response. We hypothesised that an accessible, affordable model of CRC that combines multiple cell types will improve research translation to the clinic and enable the identification of novel therapeutic targets. A viable gelatine-methacrloyl-based hydrogel culture system that incorporates CRC cells with MSCs and a monocyte cell line was developed. Gels were analysed on day 10 by PCR, cytokine array, microscopy and flow cytometry. The addition of stromal cells increased transcription of matrix remodelling proteins FN1 and MMP9, induced release of tumour-promoting immune molecules MIF, Serpin E1, CXCL1, IL-8 and CXCL12 and altered cancer cell expression of immunotherapeutic targets EGFR, CD47 and PD-L1. Treatment with PD153035, an EGFR inhibitor, revealed altered CRC expression of PD-L1 but only in gels lacking MSCs. We established a viable 3D model of CRC that combined cancer cells, MSCs and monocytic cells that can be used to research the role the stroma plays in the TME, identify novel therapeutic targets and improve the transitional efficacy of therapies.
Collapse
Affiliation(s)
- Niamh A. Leonard
- Lambe Institute for Translational Research, School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, H91 V4AY Galway, Ireland;
- Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, H91 W2TY Galway, Ireland
- Discipline of Pharmacology and Therapeutics, School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, H91 W2TY Galway, Ireland
- Correspondence: (N.A.L.); (A.E.R.)
| | - Eileen Reidy
- Lambe Institute for Translational Research, School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, H91 V4AY Galway, Ireland;
- Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, H91 W2TY Galway, Ireland
- Discipline of Pharmacology and Therapeutics, School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, H91 W2TY Galway, Ireland
| | - Kerry Thompson
- Centre for Microscopy and Imaging, Anatomy, School of Medicine, National University of Ireland Galway, H91 W2TY Galway, Ireland; (K.T.); (E.M.)
| | - Emma McDermott
- Centre for Microscopy and Imaging, Anatomy, School of Medicine, National University of Ireland Galway, H91 W2TY Galway, Ireland; (K.T.); (E.M.)
| | - Eleonora Peerani
- Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK; (E.P.); (E.T.B.); (F.R.B.); (D.L.)
| | - Elena Tomas Bort
- Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK; (E.P.); (E.T.B.); (F.R.B.); (D.L.)
| | - Frances R. Balkwill
- Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK; (E.P.); (E.T.B.); (F.R.B.); (D.L.)
| | - Daniela Loessner
- Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK; (E.P.); (E.T.B.); (F.R.B.); (D.L.)
- Faculty of Engineering and Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC 3800, Australia
- Leibniz-Institut für Polymerforschung Dresden e.V., 01069 Dresden, Germany
| | - Aideen E. Ryan
- Lambe Institute for Translational Research, School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, H91 V4AY Galway, Ireland;
- Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, H91 W2TY Galway, Ireland
- Discipline of Pharmacology and Therapeutics, School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, H91 W2TY Galway, Ireland
- Correspondence: (N.A.L.); (A.E.R.)
| |
Collapse
|
34
|
Multi-layered proteogenomic analysis unravels cancer metastasis directed by MMP-2 and focal adhesion kinase signaling. Sci Rep 2021; 11:17130. [PMID: 34429501 PMCID: PMC8385024 DOI: 10.1038/s41598-021-96635-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 08/10/2021] [Indexed: 12/27/2022] Open
Abstract
The role of matrix metalloproteinase-2 (MMP-2) in tumor cell migration has been widely studied, however, the characteristics and effects of MMP-2 in clinical sample of metastatic colorectal cancer (CRC) remain poorly understood. Here, in order to unveil the perturbed proteomic signal during MMP-2 induced cancer progression, we analyzed plasma proteome of CRC patients according to disease progression, HCT116 cancer secretome upon MMP-2 knockdown, and publicly available CRC tissue proteome data. Collectively, the integrative analysis of multi-layered proteomes revealed that a protein cluster containing EMT (Epithelial-to-Mesenchymal Transition)-associated proteins such as CD9-integrin as well as MMP-2. The proteins of the cluster were regulated by MMP-2 perturbation and exhibited significantly increased expressions in tissue and plasma as disease progressed from TNM (Tumor, Node, and Metastasis) stage I to II. Furthermore, we also identified a plausible association between MMP-2 up-regulation and activation of focal adhesion kinase signaling in the proteogenomic analysis of CRC patient tissues. Based on these comparative and integrative analyses, we suggest that the high invasiveness in the metastatic CRC resulted from increased secretion of MMP-2 and CD9-integrin complex mediated by FAK signaling activation.
Collapse
|
35
|
Carriere P, Calvo N, Novoa Díaz MB, Lopez-Moncada F, Herrera A, Torres MJ, Alonso E, Gandini NA, Gigola G, Contreras HR, Gentili C. Role of SPARC in the epithelial-mesenchymal transition induced by PTHrP in human colon cancer cells. Mol Cell Endocrinol 2021; 530:111253. [PMID: 33781836 DOI: 10.1016/j.mce.2021.111253] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 02/27/2021] [Accepted: 03/19/2021] [Indexed: 12/12/2022]
Abstract
Parathyroid hormone-related peptide (PTHrP) exerts its effects on cells derived from colorectal cancer (CRC) and tumor microenvironment and is involved in processes requiring the epithelial-mesenchymal transition (EMT). Here, we report that PTHrP modulates factors expression and morphological changes associated with EMT in HCT116 cells from CRC. PTHrP increased the protein expression of SPARC, a factor involved in EMT, in HCT116 cells but not in Caco-2 cells also from CRC but with less aggressiveness. PTHrP also increased SPARC expression and its subsequent release from endothelial HMEC-1 cells. The conditioned media of PTHrP-treated HMEC-1 cells induced early changes related to EMT in HCT116 cells. Moreover, SPARC treatment on HCT116 cells potentiated PTHrP modulation in E-cadherin expression and cell migration. In vivo PTHrP also increased SPARC expression and decreased E-cadherin expression. These results suggest a novel PTHrP action on CRC progression involving the microenvironment in the modulation of events associated with EMT.
Collapse
Affiliation(s)
- Pedro Carriere
- Department of Biology, Biochemistry and Pharmacy-INBIOSUR, National University of the South, Bahía Blanca, Argentina
| | - Natalia Calvo
- Department of Biology, Biochemistry and Pharmacy-INBIOSUR, National University of the South, Bahía Blanca, Argentina
| | - María Belén Novoa Díaz
- Department of Biology, Biochemistry and Pharmacy-INBIOSUR, National University of the South, Bahía Blanca, Argentina
| | - Fernanda Lopez-Moncada
- Department of Basic and Clinic Oncology. Faculty of Medicine, University of Chile, Chile
| | - Alexander Herrera
- Department of Basic and Clinic Oncology. Faculty of Medicine, University of Chile, Chile
| | - María José Torres
- Department of Basic and Clinic Oncology. Faculty of Medicine, University of Chile, Chile
| | | | | | - Graciela Gigola
- Department of Biology, Biochemistry and Pharmacy-INBIOSUR, National University of the South, Bahía Blanca, Argentina
| | - Hector R Contreras
- Department of Basic and Clinic Oncology. Faculty of Medicine, University of Chile, Chile
| | - Claudia Gentili
- Department of Biology, Biochemistry and Pharmacy-INBIOSUR, National University of the South, Bahía Blanca, Argentina.
| |
Collapse
|
36
|
Helmer RA, Martinez-Zaguilan R, Kaur G, Smith LA, Dufour JM, Chilton BS. Helicase-like transcription factor-deletion from the tumor microenvironment in a cell line-derived xenograft model of colorectal cancer reprogrammed the human transcriptome-S-nitroso-proteome to promote inflammation and redirect metastasis. PLoS One 2021; 16:e0251132. [PMID: 34010296 PMCID: PMC8133447 DOI: 10.1371/journal.pone.0251132] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 04/19/2021] [Indexed: 02/07/2023] Open
Abstract
Methylation of the HLTF gene in colorectal cancer (CRC) cells occurs more frequently in men than women. Progressive epigenetic silencing of HLTF in tumor cells is accompanied by negligible expression in the tumor microenvironment (TME). Cell line-derived xenografts (CDX) were established in control (Hltf+/+) and Hltf-deleted male Rag2-/-IL2rg-/- mice by direct orthotopic cell microinjection (OCMI) of HLTF+/+HCT116 Red-FLuc cells into the submucosa of the cecum. Combinatorial induction of IL6 and S100A8/A9 in the Hltf-deleted TME with ICAM-1 and IL8 in the primary tumor activated a positive feedback loop. The proinflammatory niche produced a major shift in CDX metastasis to peritoneal dissemination compared to controls. Inducible nitric oxide (iNOS) gene expression and transactivation of the iNOS-S100A8/A9 signaling complex in Hltf-deleted TME reprogrammed the human S-nitroso-proteome. POTEE, TRIM52 and UN45B were S-nitrosylated on the conserved I/L-X-C-X2-D/E motif indicative of iNOS-S100A8/A9-mediated S-nitrosylation. 2D-DIGE and protein identification by MALDI-TOF/TOF mass spectrometry authenticated S-nitrosylation of 53 individual cysteines in half-site motifs (I/L-X-C or C-X-X-D/E) in CDX tumors. POTEE in CDX tumors is both a general S-nitrosylation target and an iNOS-S100A8/A9 site-specific (Cys638) target in the Hltf-deleted TME. REL is an example of convergence of transcriptomic-S-nitroso-proteomic signaling. The gene is transcriptionally activated in CDX tumors with an Hltf-deleted TME, and REL-SNO (Cys143) was found in primary CDX tumors and all metastatic sites. Primary CDX tumors from Hltf-deleted TME shared 60% of their S-nitroso-proteome with all metastatic sites. Forty percent of SNO-proteins from primary CDX tumors were variably expressed at metastatic sites. Global S-nitrosylation of proteins in pathways related to cytoskeleton and motility was strongly implicated in the metastatic dissemination of CDX tumors. Hltf-deletion from the TME played a major role in the pathogenesis of inflammation and linked protein S-nitrosylation in primary CDX tumors with spatiotemporal continuity in metastatic progression when the tumor cells expressed HLTF.
Collapse
Affiliation(s)
- Rebecca A. Helmer
- Department of Cell Biology & Biochemistry, Texas Tech University Health Sciences Center, Lubbock, Texas, United States of America
| | - Raul Martinez-Zaguilan
- Department of Cell Physiology & Molecular Biophysics, Texas Tech University Health Sciences Center, Lubbock, Texas, United States of America
| | - Gurvinder Kaur
- Department of Medical Education, Texas Tech University Health Sciences Center, Lubbock, Texas, United States of America
| | - Lisa A. Smith
- Department of Pathology, Texas Tech University Health Sciences Center, Lubbock, Texas, United States of America
| | - Jannette M. Dufour
- Department of Cell Biology & Biochemistry, Texas Tech University Health Sciences Center, Lubbock, Texas, United States of America
| | - Beverly S. Chilton
- Department of Cell Biology & Biochemistry, Texas Tech University Health Sciences Center, Lubbock, Texas, United States of America
- Cancer Center, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, United States of America
| |
Collapse
|
37
|
Martens MC, Edelkamp J, Seebode C, Schäfer M, Stählke S, Krohn S, Jung O, Murua Escobar H, Emmert S, Boeckmann L. Generation and Characterization of a CRISPR/Cas9-Mediated SNAP29 Knockout in Human Fibroblasts. Int J Mol Sci 2021; 22:ijms22105293. [PMID: 34069872 PMCID: PMC8157373 DOI: 10.3390/ijms22105293] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/07/2021] [Accepted: 05/11/2021] [Indexed: 12/26/2022] Open
Abstract
Loss-of-function mutations in the synaptosomal-associated protein 29 (SNAP29) lead to the rare autosomal recessive neurocutaneous cerebral dysgenesis, neuropathy, ichthyosis, and keratoderma (CEDNIK) syndrome. SNAP29 is a soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) protein. So far, it has been shown to be involved in membrane fusion, epidermal differentiation, formation of primary cilia, and autophagy. Recently, we reported the successful generation of two mouse models for the human CEDNIK syndrome. The aim of this investigation was the generation of a CRISPR/Cas9-mediated SNAP29 knockout (KO) in an immortalized human cell line to further investigate the role of SNAP29 in cellular homeostasis and signaling in humans independently of animal models. Comparison of different methods of delivery for CRISPR/Cas9 plasmids into the cell revealed that lentiviral transduction is more efficient than transfection methods. Here, we reported to the best of our knowledge the first successful generation of a CRISPR/Cas9-mediated SNAP29 KO in immortalized human MRC5Vi fibroblasts (c.169_196delinsTTCGT) via lentiviral transduction.
Collapse
Affiliation(s)
- Marie Christine Martens
- Clinic and Policlinic for Dermatology and Venerology, University Medical Center Rostock, 18057 Rostock, Germany; (M.C.M.); (J.E.); (C.S.); (M.S.); (O.J.); (S.E.)
| | - Janin Edelkamp
- Clinic and Policlinic for Dermatology and Venerology, University Medical Center Rostock, 18057 Rostock, Germany; (M.C.M.); (J.E.); (C.S.); (M.S.); (O.J.); (S.E.)
| | - Christina Seebode
- Clinic and Policlinic for Dermatology and Venerology, University Medical Center Rostock, 18057 Rostock, Germany; (M.C.M.); (J.E.); (C.S.); (M.S.); (O.J.); (S.E.)
| | - Mirijam Schäfer
- Clinic and Policlinic for Dermatology and Venerology, University Medical Center Rostock, 18057 Rostock, Germany; (M.C.M.); (J.E.); (C.S.); (M.S.); (O.J.); (S.E.)
| | - Susanne Stählke
- Department of Cell Biology, University Medical Center Rostock, 18057 Rostock, Germany;
| | - Saskia Krohn
- Clinic for Hematology, Oncology and Palliative Care, University Medical Center Rostock, 18057 Rostock, Germany; (S.K.); (H.M.E.)
| | - Ole Jung
- Clinic and Policlinic for Dermatology and Venerology, University Medical Center Rostock, 18057 Rostock, Germany; (M.C.M.); (J.E.); (C.S.); (M.S.); (O.J.); (S.E.)
| | - Hugo Murua Escobar
- Clinic for Hematology, Oncology and Palliative Care, University Medical Center Rostock, 18057 Rostock, Germany; (S.K.); (H.M.E.)
| | - Steffen Emmert
- Clinic and Policlinic for Dermatology and Venerology, University Medical Center Rostock, 18057 Rostock, Germany; (M.C.M.); (J.E.); (C.S.); (M.S.); (O.J.); (S.E.)
| | - Lars Boeckmann
- Clinic and Policlinic for Dermatology and Venerology, University Medical Center Rostock, 18057 Rostock, Germany; (M.C.M.); (J.E.); (C.S.); (M.S.); (O.J.); (S.E.)
- Correspondence:
| |
Collapse
|
38
|
Quantitative Proteomics Analysis of Berberine-Treated Colon Cancer Cells Reveals Potential Therapy Targets. BIOLOGY 2021; 10:biology10030250. [PMID: 33806918 PMCID: PMC8005188 DOI: 10.3390/biology10030250] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/13/2021] [Accepted: 03/20/2021] [Indexed: 02/07/2023]
Abstract
Simple Summary Colon cancer is one of the most common malignant tumors and beberine has been found to exert potential anti-colon cancer activity in vitro and in vivo. In this study, by using proteomics and bioinformatics approaches, we report that berberine may inhibit the proliferation of colon cancer cells by regulating mitochondrial translation and ribosome biogenesis, as well as by promoting calcium mobilization and metabolism of fat-soluble vitamins. Moreover, GTPase ERAL1 and mitochondrial ribosomal proteins MRPL11, 15, 30, 37, 40, and 52 have great potential to serve as potential therapeutic targets for colon cancer treatment. Abstract Colon cancer is one of the most lethal malignancies worldwide. Berberine has been found to exert potential anti-colon cancer activity in vitro and in vivo, although the detailed regulatory mechanism is still unclear. This study aims to identify the underlying crucial proteins and regulatory networks associated with berberine treatment of colon cancer by using proteomics as well as publicly available transcriptomics and tissue array data. Proteome profiling of berberine-treated colon cancer cells demonstrated that among 5130 identified proteins, the expression of 865 and 675 proteins were changed in berberine-treated HCT116 and DLD1 cells, respectively. Moreover, 54 differently expressed proteins that overlapped in both cell lines were mainly involved in mitochondrial protein synthesis, calcium mobilization, and metabolism of fat-soluble vitamins. Finally, GTPase ERAL1 and mitochondrial ribosomal proteins including MRPL11, 15, 30, 37, 40, and 52 were identified as hub proteins of berberine-treated colon cancer cells. These proteins have higher transcriptional and translational levels in colon tumor samples than that of colon normal samples, and were significantly down-regulated in berberine-treated colon cancer cells. Genetic dependency analysis showed that silencing the gene expression of seven hub proteins could inhibit the proliferation of colon cancer cells. This study sheds a light for elucidating the berberine-related regulatory signaling pathways in colon cancer, and suggests that ERAL1 and several mitochondrial ribosomal proteins might be promising therapeutic targets for colon cancer.
Collapse
|
39
|
Hollenbeck MG, Blevins AA, Gatrone EE, Hundal T, O'Connell K, Lavigne JJ. Identifying Synthetic Lectins from a Competitive Screen for the Detection of Prostate Cancer. Bioconjug Chem 2020; 31:2750-2758. [PMID: 33275847 DOI: 10.1021/acs.bioconjchem.0c00526] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Herein, we report a dual dye competitive screening method for the identification of five boronic acid functionalized synthetic lectins (SLs) that are selective for prostate-associated targets with the goal of detecting and staging prostate cancer. This method uses differently labeled normal (RWEP-1) and diseased (PC3) cell membrane extracts in a competitive binding assay to identify SLs that bind either the cancerous or normal extracts but not both. Subsequent studies examined the efficacy of these new SL hits in an array format to discriminate six prostate cell lines. The SL array was able to (a) classify the prostate cell lines with 83% accuracy, (b) discriminate the same cell lines based on their metastatic potential (noncancerous/healthy, cancerous/lowly metastatic, and cancerous/metastatic) with 96% classification accuracy, and (c) exhibit enhanced selectivity for prostate-derived versus colon-derived cell lines. Further analysis delineated the contribution from each SL in these studies, providing a focused SL array having potential utility as a cancer diagnostic.
Collapse
Affiliation(s)
- Mary Grace Hollenbeck
- Department of Chemistry & Biochemistry, University of South Carolina, 631 Sumter Street, Columbia, South Carolina 29208, United States
| | - Anna A Blevins
- Department of Chemistry & Biochemistry, University of South Carolina, 631 Sumter Street, Columbia, South Carolina 29208, United States
| | - Erin E Gatrone
- Department of Chemistry & Biochemistry, University of South Carolina, 631 Sumter Street, Columbia, South Carolina 29208, United States
| | - Tanya Hundal
- Department of Chemistry & Biochemistry, University of South Carolina, 631 Sumter Street, Columbia, South Carolina 29208, United States
| | - Kathleen O'Connell
- Department of Chemistry & Biochemistry, University of South Carolina, 631 Sumter Street, Columbia, South Carolina 29208, United States
| | - John J Lavigne
- Department of Chemistry & Biochemistry, University of South Carolina, 631 Sumter Street, Columbia, South Carolina 29208, United States
| |
Collapse
|
40
|
Wang X, Saud SM, Wang F, He S, Zhang X, Hua B, Li W. Protective effect of ShaoYao decoction on colitis-associated colorectal cancer by inducing Nrf2 signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2020; 252:112600. [PMID: 31981745 DOI: 10.1016/j.jep.2020.112600] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/10/2020] [Accepted: 01/18/2020] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ulcerative colitis were prone to develop into ulcerrelated colorectal cancer with high risk of mortality. Shaoyao Decoction (SYD), a compound prescription of Chinese traditional medicine, was reported to have anti-colorectal cancer effect. Thus this study mainly investigated the protective and preventive effect of SYD against oxidative damages and inflamatory response through in vivo and in vitro experiments. AIM OF THE STUDY Shaoyao decoction (SYD), a compound prescription of traditional Chinese medicine, is effective in treating ulcerative colitis. The increased levels of reactive oxygen species (ROS) in inflammatory cells potentially drive the development of carcinomas. Nuclear factor-erythroid 2-related factor 2 (Nrf2) has became a novel target for the prevention of colorectal cancer (CRC). In this study, we assessed the antioxidation effect of SYD against colitis associated colorectal cancer through in vivo and in vitro experiments. MATERIALS AND METHODS In vivo AOM/DSS-induced murine model of colon cancer and in vitro H2O2-induced oxidative stress in HT-29 cells model were conducted. To determine the antioxidant activity of SYD, protein expression of Nrf2 and its downstream genes were detected by western blot, RT-PCR and Enzyme-linked immunosorbent assay. RESULTS Both in vivo and in vitro experiments demonstrated that SYD exerts antioxidant effect through activation of Nrf2 pathway and upregulation expression of Nrf2 downstream genes. SYD is shown to have preventive effect against colitis-associated colorectal cancer. CONCLUSIONS These observations suggest that SYD is effective in the enhancement of antioxidant ability via activation of Nrf2 pathway and the up-regulation of Nrf2-downstream phase II enzymes expression. The anti-inflammation and antioxidant action of SYD together contributes to the prevention and treatment of ulcerrelated colorectal cancer.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5, Beixiange Road, Xicheng District, Beijing, 100053, China; Department of Medical Security Management, Wangjing Hospital, China Academy of Chinese Medical Sciences, No.6, Zhonghuan South Road, Chaoyang District, Beijing, 100102, China
| | - Shakir M Saud
- Department of Family Medicine, Contra Costa Regional Medical Center, Martinez, CA, 94553, USA
| | - Fang Wang
- Department of Oncology, Wangjing Hospital, China Academy of Chinese Medical Sciences, No.6, Huajiadi Road, Chaoyang District, Beijing, 100102, China
| | - Shengqi He
- Department of Oncology, Wangjing Hospital, China Academy of Chinese Medical Sciences, No.6, Huajiadi Road, Chaoyang District, Beijing, 100102, China
| | - Xiwen Zhang
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5, Beixiange Road, Xicheng District, Beijing, 100053, China
| | - Baojin Hua
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5, Beixiange Road, Xicheng District, Beijing, 100053, China.
| | - Weidong Li
- Department of Scientific Research Management, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5, Beixiange Road, Xicheng District, Beijing, 100053, China.
| |
Collapse
|
41
|
Nair-Menon J, Daulagala AC, Connor DM, Rutledge L, Penix T, Bridges MC, Wellslager B, Spyropoulos DD, Timmers CD, Broome AM, Kourtidis A. Predominant Distribution of the RNAi Machinery at Apical Adherens Junctions in Colonic Epithelia Is Disrupted in Cancer. Int J Mol Sci 2020; 21:E2559. [PMID: 32272708 PMCID: PMC7177752 DOI: 10.3390/ijms21072559] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/04/2020] [Accepted: 04/04/2020] [Indexed: 12/30/2022] Open
Abstract
The RNA interference (RNAi) machinery is an essential component of the cell, regulating miRNA biogenesis and function. RNAi complexes were thought to localize either in the nucleus, such as the microprocessor, or in the cytoplasm, such as the RNA-induced silencing complex (RISC). We recently revealed that the core microprocessor components DROSHA and DGCR8, as well as the main components of RISC, including Ago2, also associate with the apical adherens junctions of well-differentiated cultured epithelial cells. Here, we demonstrate that the localization of the core RNAi components is specific and predominant at apical areas of cell-cell contact of human normal colon epithelial tissues and normal primary colon epithelial cells. Importantly, the apical junctional localization of RNAi proteins is disrupted or lost in human colon tumors and in poorly differentiated colon cancer cell lines, correlating with the dysregulation of the adherens junction component PLEKHA7. We show that the restoration of PLEKHA7 expression at adherens junctions of aggressively tumorigenic colon cancer cells restores the junctional localization of RNAi components and suppresses cancer cell growth in vitro and in vivo. In summary, this work identifies the apical junctional localization of the RNAi machinery as a key feature of the differentiated colonic epithelium, with a putative tumor suppressing function.
Collapse
Affiliation(s)
- Joyce Nair-Menon
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA; (J.N.-M.); (A.C.D.); (L.R.); (T.P.); (M.C.B.); (B.W.)
| | - Amanda C. Daulagala
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA; (J.N.-M.); (A.C.D.); (L.R.); (T.P.); (M.C.B.); (B.W.)
| | - Dean M. Connor
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA; (D.M.C.); (A.-M.B.)
| | - Lauren Rutledge
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA; (J.N.-M.); (A.C.D.); (L.R.); (T.P.); (M.C.B.); (B.W.)
| | - Trevor Penix
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA; (J.N.-M.); (A.C.D.); (L.R.); (T.P.); (M.C.B.); (B.W.)
| | - Mary Catherine Bridges
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA; (J.N.-M.); (A.C.D.); (L.R.); (T.P.); (M.C.B.); (B.W.)
| | - Bridgette Wellslager
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA; (J.N.-M.); (A.C.D.); (L.R.); (T.P.); (M.C.B.); (B.W.)
| | - Demetri D. Spyropoulos
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA;
| | - Cynthia D. Timmers
- Department of Medicine, Medical University of South Carolina, 171 Ashley Avenue, Charleston, SC 29425, USA;
| | - Ann-Marie Broome
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA; (D.M.C.); (A.-M.B.)
| | - Antonis Kourtidis
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA; (J.N.-M.); (A.C.D.); (L.R.); (T.P.); (M.C.B.); (B.W.)
| |
Collapse
|
42
|
Silva FL, Silva RV, Branco PC, Costa-Lotufo LV, Murakami C, Young MC, Azevedo DA, Moreno PR. Chemical composition of the Brazilian native Cinnamomum stenophyllum (Meisn.) Vattimo-Gil essential oil by GC-qMS and GC × GC-TOFMS, and its cytotoxic activity. ARAB J CHEM 2020. [DOI: 10.1016/j.arabjc.2020.01.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
43
|
Xu Y, Zhang L, Wang Q, Zheng M. Comparison of Different Colorectal Cancer With Liver Metastases Models Using Six Colorectal Cancer Cell Lines. Pathol Oncol Res 2020; 26:2177-2183. [PMID: 32172478 DOI: 10.1007/s12253-020-00805-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 03/04/2020] [Indexed: 02/06/2023]
Abstract
At present, modeling methods of colorectal cancer with liver metastases have significant limitations. Here, we established orthotopic and ectopic hepatic metastases models using six colorectal cancer cell lines to choose an ideal animal model for studying colorectal cancer growth and liver metastases. Luciferin-expressing six colorectal cancer cell lines were used to induce animal models of colorectal cancer with liver metastases by intra-splenic injection or implantation of tumor tissue in the caecum. Tumors growth and metastatic events were observed by bioluminescence imaging. In orthotopic transplantation group, six cell lines all had taken rates of 100% for orthotopic tumors but showed variations in rates of growth. HCT-116 cell developed the 50% liver metastases. However, the ectopic transplantation group achieved higher liver metastatic rate, with the highest frequencies for HCT116 cell (90%) and SW620 cell (77.8%). Furthermore, the time to develop liver metastases and survival rates of bearing-tumor mice were shorter than orthotopic transplantation group. Additionally, six colorectal cancer cell lines resulted in more lymph node metastases in orthotopic transplantation group, whereas produced widespread peritoneal seeding in ectopic transplantation group. Bioluminescence imaging and pathological findings confirmed the growth and metastatic characteristics of tumors. Two animal models of colorectal cancer using six cell lines showed highly variations in rates of growth, survival rates of bearing-tumor mice and frequencies of metastases. The study provides useful information for the establishment of clinically relevant colorectal cancer with liver metastases animal models.
Collapse
Affiliation(s)
- Yuting Xu
- Department of Pathology, Xuzhou Medical University, 221004, Xuzhou, Jiangsu, China.
| | - Lin Zhang
- Department of Pathology, Xuzhou Medical University, 221004, Xuzhou, Jiangsu, China
| | - Qingling Wang
- Department of Pathology, Xuzhou Medical University, 221004, Xuzhou, Jiangsu, China
| | - Maojin Zheng
- Department of Pathology, Xuzhou Medical University, 221004, Xuzhou, Jiangsu, China
| |
Collapse
|
44
|
Liao C, Li M, Li X, Li N, Zhao X, Wang X, Song Y, Quan J, Cheng C, Liu J, Bode AM, Cao Y, Luo X. Trichothecin inhibits invasion and metastasis of colon carcinoma associating with SCD-1-mediated metabolite alteration. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158540. [PMID: 31678511 DOI: 10.1016/j.bbalip.2019.158540] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 10/16/2019] [Accepted: 10/17/2019] [Indexed: 02/08/2023]
Abstract
Lipid metabolic abnormalities have received intensified concerns and increased de novo synthesis of lipids is recognized as a common feature of many human cancers. Nevertheless, the role of lipid metabolism that confers aggressive properties on human cancers still remains to be revealed. Natural compounds represent an abundant pool of agents for the discovery of novel lead compounds. Trichothecin (TCN) is a sesquiterpenoid originating from an endophytic fungus of the herbal plant Maytenus hookeri Loes. Here, we assess the association of stearoyl-CoA desaturase 1 (SCD-1) over-expression with malignant progression of colorectal cancer (CRC). Based on this association, the effect of TCN on migration and invasion of colon carcinoma cells closely related to the inhibition of SCD-1 is evaluated. We further demonstrate that reduced production of unsaturated fatty acids (FAs) by blocking SCD-1 activity is beneficial for the anti-invasion effect of TCN. The aim of this study was to clarify the mechanistic connection between metabolite alterations induced by metabolic rewiring and the aggressive tumor phenotype and further develop novel pharmacological tools for the intervention of tumor invasion associated with SCD-1-mediated metabolite alterations.
Collapse
Affiliation(s)
- Chaoliang Liao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan 410078, China; Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, China; Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Changsha, Hunan 410078, China
| | - Min Li
- Department of Oncology, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210001, China
| | - Xiang Li
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan 410078, China
| | - Namei Li
- Key Laboratory of Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan 410078, China; Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, China; Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Changsha, Hunan 410078, China
| | - Xu Zhao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan 410078, China; Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, China; Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Changsha, Hunan 410078, China
| | - Xiaoyi Wang
- Key Laboratory of Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan 410078, China; Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, China; Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Changsha, Hunan 410078, China
| | - Yawen Song
- Key Laboratory of Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan 410078, China; Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, China; Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Changsha, Hunan 410078, China
| | - Jing Quan
- Key Laboratory of Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan 410078, China; Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, China; Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Changsha, Hunan 410078, China
| | - Can Cheng
- Key Laboratory of Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan 410078, China; Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, China; Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Changsha, Hunan 410078, China
| | - Jikai Liu
- School of Pharmacy, South-central University for Nationalities, Wuhan, Hubei 430074, China
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Ya Cao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan 410078, China; Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, China; Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Changsha, Hunan 410078, China; Molecular Imaging Research Center of Central South University, Changsha 410008, Hunan, China; National Joint Engineering Research Center for Genetic Diagnostics of Infectious Diseases and Cancer, Changsha 410078, China
| | - Xiangjian Luo
- Key Laboratory of Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan 410078, China; Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, China; Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Changsha, Hunan 410078, China; Molecular Imaging Research Center of Central South University, Changsha 410008, Hunan, China.
| |
Collapse
|
45
|
Kim JS, Lee D, Kim D, Mun SJ, Cho E, Son W, Yang CS. Toxoplasma gondii GRA8-derived peptide immunotherapy improves tumor targeting of colorectal cancer. Oncotarget 2020; 11:62-73. [PMID: 32002124 PMCID: PMC6967779 DOI: 10.18632/oncotarget.27417] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 12/19/2019] [Indexed: 12/20/2022] Open
Abstract
Targeted tumor and efficient, specific biological drug delivery in vivo has been one of the main challenges in protein-based cancer-targeted therapies. Mitochondria are potential therapeutic targets for various anti-cancer drugs. We have previously reported that protein kinase Cα-mediated phosphorylation of Toxoplasma gondii GRA8 is required for mitochondrial trafficking and regulating the interaction of the C-terminal of GRA8 with ATP5A1/SIRT3 in mitochondria. Furthermore, SIRT3 facilitates ATP5A1 deacetylation, mitochondrial activation, and subsequent antiseptic activity in vivo. Herein we developed a recombinant acidity-triggered rational membrane (ATRAM)-conjugated multifunctional GRA8 peptide (rATRAM-G8-M/AS) comprising ATRAM as the cancer-targeting cell-penetrating peptide, and essential/minimal residues for mitochondrial targeting or ATP5A1/SIRT3 binding. This peptide construct showed considerably improved potency about cancer cell death via mitochondria activity and biogenesis compared with rGRA8 alone in HCT116 human carcinoma cells, reaching an IC50 value of up to 200-fold lower in vitro and 500-fold lower in vivo. Notably, rATRAM-G8-M/AS treatment showed significant therapeutic effects in a mouse xenograft model through mitochondrial metabolic resuscitation, and it produced negligible immunogenicity and immune responses in vivo. Thus, these results demonstrate that rATRAM-G8-M/AS represents a useful therapeutic strategy against tumors, particularly colon cancer. This strategy represents an urgently needed paradigm shift for therapeutic intervention.
Collapse
Affiliation(s)
- Jae-Sung Kim
- Department of Molecular and Life Science, Hanyang University, Ansan 15588, S. Korea.,Department of Bionano Technology, Hanyang University, Seoul 04673, S. Korea
| | - Daeun Lee
- Department of Molecular and Life Science, Hanyang University, Ansan 15588, S. Korea
| | - Donggyu Kim
- Department of Molecular and Life Science, Hanyang University, Ansan 15588, S. Korea
| | - Seok-Jun Mun
- Department of Molecular and Life Science, Hanyang University, Ansan 15588, S. Korea.,Department of Bionano Technology, Hanyang University, Seoul 04673, S. Korea
| | - Euni Cho
- Department of Molecular and Life Science, Hanyang University, Ansan 15588, S. Korea.,Department of Bionano Technology, Hanyang University, Seoul 04673, S. Korea
| | - Wooic Son
- Department of Molecular and Life Science, Hanyang University, Ansan 15588, S. Korea
| | - Chul-Su Yang
- Department of Molecular and Life Science, Hanyang University, Ansan 15588, S. Korea.,Department of Bionano Technology, Hanyang University, Seoul 04673, S. Korea
| |
Collapse
|
46
|
Caratelli S, Arriga R, Sconocchia T, Ottaviani A, Lanzilli G, Pastore D, Cenciarelli C, Venditti A, Del Principe MI, Lauro D, Landoni E, Du H, Savoldo B, Ferrone S, Dotti G, Sconocchia G. In vitro elimination of epidermal growth factor receptor-overexpressing cancer cells by CD32A-chimeric receptor T cells in combination with cetuximab or panitumumab. Int J Cancer 2020; 146:236-247. [PMID: 31479522 PMCID: PMC8711771 DOI: 10.1002/ijc.32663] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 08/21/2019] [Accepted: 08/23/2019] [Indexed: 02/03/2023]
Abstract
Cetuximab and panitumumab bind the human epidermal growth factor receptor (EGFR). Although the chimeric cetuximab (IgG1) triggers antibody-dependent-cellular-cytotoxicity (ADCC) of EGFR positive target cells, panitumumab (a human IgG2) does not. The inability of panitumumab to trigger ADCC reflects the poor binding affinity of human IgG2 Fc for the FcγRIII (CD16) on natural killer (NK) cells. However, both human IgG1 and IgG2 bind the FcγRII (CD32A) to a similar extent. Our study compares the ability of T cells, engineered with a novel low-affinity CD32A131R -chimeric receptor (CR), and those engineered with the low-affinity CD16158F -CR T cells, in eliminating EGFR positive epithelial cancer cells (ECCs) in combination with cetuximab or panitumumab. After T-cell transduction, the percentage of CD32A131R -CR T cells was 74 ± 10%, whereas the percentage of CD16158F -CR T cells was 46 ± 15%. Only CD32A131R -CR T cells bound panitumumab. CD32A131R -CR T cells combined with the mAb 8.26 (anti-CD32) and CD16158F -CR T cells combined with the mAb 3g8 (anti-CD16) eliminated colorectal carcinoma (CRC), HCT116FcγR+ cells, in a reverse ADCC assay in vitro. Crosslinking of CD32A131R -CR on T cells by cetuximab or panitumumab and CD16158F -CR T cells by cetuximab induced elimination of triple negative breast cancer (TNBC) MDA-MB-468 cells, and the secretion of interferon gamma and tumor necrosis factor alpha. Neither cetuximab nor panitumumab induced Fcγ-CR T antitumor activity against Kirsten rat sarcoma (KRAS)-mutated HCT116, nonsmall-cell-lung-cancer, A549 and TNBC, MDA-MB-231 cells. The ADCC of Fcγ-CR T cells was associated with the overexpression of EGFR on ECCs. In conclusion, CD32A131R -CR T cells are efficiently redirected by cetuximab or panitumumab against breast cancer cells overexpressing EGFR.
Collapse
Affiliation(s)
- Sara Caratelli
- Department of Biomedical Sciences, Institute of Translational Pharmacology, CNR, Rome, Italy
| | - Roberto Arriga
- Department of Systems Medicine, University of Rome “Tor Vergata”, Rome, Italy
| | - Tommaso Sconocchia
- Otto Loewi Research Center, Chair of Immunology and Pathophysiology, Medical University of Graz, Graz, Austria
| | - Alessio Ottaviani
- Department of Biomedical Sciences, Institute of Translational Pharmacology, CNR, Rome, Italy
| | - Giulia Lanzilli
- Department of Biomedical Sciences, Institute of Translational Pharmacology, CNR, Rome, Italy
| | - Donatella Pastore
- Department of Systems Medicine, University of Rome “Tor Vergata”, Rome, Italy
| | - Carlo Cenciarelli
- Department of Biomedical Sciences, Institute of Translational Pharmacology, CNR, Rome, Italy
| | - Adriano Venditti
- Hematology, Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Rome, Italy
| | | | - Davide Lauro
- Department of Systems Medicine, University of Rome “Tor Vergata”, Rome, Italy
| | - Elisa Landoni
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| | - Hongwei Du
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| | - Barbara Savoldo
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| | - Soldano Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Gianpietro Dotti
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| | - Giuseppe Sconocchia
- Department of Biomedical Sciences, Institute of Translational Pharmacology, CNR, Rome, Italy
| |
Collapse
|
47
|
Leiphrakpam PD, Lazenby AJ, Chowdhury S, Smith LM, Mathiesen M, Brattain MG, Wang J, Black JD, Are C. Prognostic and therapeutic implications of NHERF1 expression and regulation in colorectal cancer. J Surg Oncol 2019; 121:547-560. [PMID: 31867736 DOI: 10.1002/jso.25805] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 11/22/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Na+ /H+ exchanger regulatory factor 1 (NHERF1) has been implicated in the tumorigenesis of several cancer types and is a potential therapeutic target. The current study evaluated the relationship between NHERF1 expression and clinical outcome in colorectal cancer (CRC). METHODS NHERF1 expression was evaluated by immunohistochemistry in 167 patients with CRC primary tumors, 37 patients with no disease, and 27 patients with metastatic CRC (mCRC); and in the orthotopically implanted tumors in mice. NHERF1 expression was manipulated in CRC cells using inducible short hairpin RNAs to determine its biological functions. RESULTS High expression of NHERF1 correlated with CRC progression and metastasis, as well as significantly worse overall survival, recurrence-free survival, and disease-specific survival. Orthotopic implantation studies demonstrated increased NHERF1 expression in liver metastases. Treatment of CRC xenografts with insulin-like growth factor 1 receptor (IGF1R) inhibitors downregulated NHERF1 expression, indicating NHERF1 is downstream of IGF1R signaling. Knockdown of NHERF1 increased apoptosis and reduced X-linked inhibitor of apoptosis protein (XIAP) and survivin expression, indicating NHERF1 is critical for CRC cell survival. CONCLUSION NHERF1 expression levels correlated with worse prognosis in patients with CRC and plays a critical role in CRC cell survival. Together, our findings establish NHERF1 as a novel potential marker for increased risk of CRC-specific mortality and identify NHERF1 as an attractive therapeutic target for mCRC treatment.
Collapse
Affiliation(s)
- Premila D Leiphrakpam
- Department of Surgery, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Audrey J Lazenby
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Sanjib Chowdhury
- Section of Gastroenterology, College of Medicine, Boston University Medical Center, Boston, Massachusetts
| | - Lynette M Smith
- Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, Omaha, Nebraska
| | - Michelle Mathiesen
- Diagnostic Laboratory, Phibro Animal Health Corporation, Omaha, Nebraska
| | - Michael G Brattain
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska
| | - Jing Wang
- Department of Cancer Biology and Genetics, College of Medicine, Ohio State University, Columbus, Ohio
| | - Jennifer D Black
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska
| | - Chandrakanth Are
- Division of Surgical Oncology, Department of Surgery, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
48
|
Sastre D, Baiochi J, de Souza Lima IM, Canto de Souza F, Corveloni AC, Thomé CH, Faça VM, Schiavinato JLDS, Covas DT, Panepucci RA. Focused screening reveals functional effects of microRNAs differentially expressed in colorectal cancer. BMC Cancer 2019; 19:1239. [PMID: 31864341 PMCID: PMC6925883 DOI: 10.1186/s12885-019-6468-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 12/16/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is still a leading cause of death worldwide. Recent studies have pointed to an important role of microRNAs in carcinogenesis. Several microRNAs are described as aberrantly expressed in CRC tissues and in the serum of patients. However, functional outcomes of microRNA aberrant expression still need to be explored at the cellular level. Here, we aimed to investigate the effects of microRNAs aberrantly expressed in CRC samples in the proliferation and cell death of a CRC cell line. METHODS We transfected 31 microRNA mimics into HCT116 cells. Total number of live propidium iodide negative (PI-) and dead (PI+) cells were measured 4 days post-transfection by using a high content screening (HCS) approach. HCS was further used to evaluate apoptosis (via Annexin V and PI staining), and to discern between intrinsic and extrinsic apoptotic pathways, by detecting cleaved Caspase 9 and 8, respectively. To reveal mRNA targets and potentially involved mechanisms, we performed microarray gene expression and functional pathway enrichment analysis. Quantitative PCR and western blot were used to validate potential mRNA targets. RESULTS Twenty microRNAs altered the proliferation of HCT116 cells in comparison to control. miR-22-3p, miR-24-3p, and miR-101-3p significantly repressed cell proliferation and induced cell death. Interestingly, all anti-proliferative microRNAs in our study had been previously described as poorly expressed in the CRC samples. Predicted miR-101-3p targets that were also downregulated by in our microarray were enriched for genes associated with Wnt and cancer pathways, including MCL-1, a member of the BCL-2 family, involved in apoptosis. Interestingly, miR-101-3p preferentially downregulated the long anti-apoptotic MCL-1 L isoform, and reduced cell survival specifically by activating the intrinsic apoptosis pathway. Moreover, miR-101-3p also downregulated IL6ST, STAT3A/B, and MYC mRNA levels, genes associated with stemness properties of CRC cells. CONCLUSIONS microRNAs upregulated in CRC tend to induce proliferation in vitro, whereas microRNAs poorly expressed in CRC halt proliferation and induce cell death. We provide novel evidence linking preferential inhibition of the anti-apoptotic MCL-1 L isoform by miR-101-3p and consequent activation of the intrinsic apoptotic pathway as potential mechanisms for its antitumoral activity, likely due to the inhibition of the IL-6/JAK/STAT signaling pathway.
Collapse
Affiliation(s)
- Danuta Sastre
- Laboratory of Human and Medical Genetics, Federal University of Pará, Rua Augusto Corrêa, 01. Guamá., Belém, Pará CEP 66075-110 Brazil
- Laboratory of Functional Biology (LFBio), Center for Cell-Based Therapy (CTC), Regional Blood Center, Ribeirao Preto Medical School, University of São Paulo (USP), R. Ten. Catão Roxo, 2501., Ribeirão Preto, SP 14051-140 Brazil
| | - João Baiochi
- Laboratory of Functional Biology (LFBio), Center for Cell-Based Therapy (CTC), Regional Blood Center, Ribeirao Preto Medical School, University of São Paulo (USP), R. Ten. Catão Roxo, 2501., Ribeirão Preto, SP 14051-140 Brazil
| | - Ildercilio Mota de Souza Lima
- Laboratory of Functional Biology (LFBio), Center for Cell-Based Therapy (CTC), Regional Blood Center, Ribeirao Preto Medical School, University of São Paulo (USP), R. Ten. Catão Roxo, 2501., Ribeirão Preto, SP 14051-140 Brazil
| | - Felipe Canto de Souza
- Laboratory of Functional Biology (LFBio), Center for Cell-Based Therapy (CTC), Regional Blood Center, Ribeirao Preto Medical School, University of São Paulo (USP), R. Ten. Catão Roxo, 2501., Ribeirão Preto, SP 14051-140 Brazil
| | - Amanda Cristina Corveloni
- Laboratory of Functional Biology (LFBio), Center for Cell-Based Therapy (CTC), Regional Blood Center, Ribeirao Preto Medical School, University of São Paulo (USP), R. Ten. Catão Roxo, 2501., Ribeirão Preto, SP 14051-140 Brazil
| | - Carolina Hassib Thomé
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo (USP), Av. Bandeirantes, 3900 - Vila Monte Alegre, Ribeirão Preto, SP 14049-900 Brazil
| | - Vitor Marcel Faça
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo (USP), Av. Bandeirantes, 3900 - Vila Monte Alegre, Ribeirão Preto, SP 14049-900 Brazil
| | - Josiane Lilian dos Santos Schiavinato
- Laboratory of Functional Biology (LFBio), Center for Cell-Based Therapy (CTC), Regional Blood Center, Ribeirao Preto Medical School, University of São Paulo (USP), R. Ten. Catão Roxo, 2501., Ribeirão Preto, SP 14051-140 Brazil
| | - Dimas Tadeu Covas
- Laboratory of Functional Biology (LFBio), Center for Cell-Based Therapy (CTC), Regional Blood Center, Ribeirao Preto Medical School, University of São Paulo (USP), R. Ten. Catão Roxo, 2501., Ribeirão Preto, SP 14051-140 Brazil
| | - Rodrigo Alexandre Panepucci
- Laboratory of Functional Biology (LFBio), Center for Cell-Based Therapy (CTC), Regional Blood Center, Ribeirao Preto Medical School, University of São Paulo (USP), R. Ten. Catão Roxo, 2501., Ribeirão Preto, SP 14051-140 Brazil
| |
Collapse
|
49
|
Wu Y, Feng Z, Jiang S, Chen J, Zhan Y, Chen J. Secreting-lux/pT-ClyA engineered bacteria suppresses tumor growth via interleukin-1β in two pathways. AMB Express 2019; 9:189. [PMID: 31754923 PMCID: PMC6872689 DOI: 10.1186/s13568-019-0910-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 11/04/2019] [Indexed: 01/21/2023] Open
Abstract
Engineered Salmonella typhimurium (S.t-ΔpGlux/pT-ClyA) and attenuated Salmonella typhimurium (SL: Salmonella typhimurium with a defect in the synthesis of guanine 5′-diphosphate-3′-diphosphate) exhibit similar tumor targeting capabilities (Kim et al. in Theranostics 5:1328–1342, 2015; Jiang et al. in Mol Ther 18:635–642, 2013), but S.t-ΔpGlux/pT-ClyA exerts superior tumor suppressive effects. The aim of this study was to investigate whether S.t-ΔpGlux/pT-ClyA inhibits colon cancer growth and recurrence by promoting increased IL-1β production. The CT26 tumor mouse model was used, and mice were treated in the following ways: PBS, S.t-ΔpGlux/pT-ClyA(+) + IL-1βAb, SL, S.t-ΔpGlux/pT-ClyA(−), and S.t-ΔpGlux/pT-ClyA(+). Dynamic evaluation of the efficacy of S.t-ΔpGlux/pT-ClyA in the treatment of colon cancer was assessed by MRI. Western blot, immunofluorescence and flow cytometry analysis were used to investigate IL-1β-derived cells and IL-1β expression on tumor cells and immune cells to analyze the regulatory mechanism. IL-1β levels in tumors colonized by S.t-ΔpGlux/pT-ClyA were significantly increased and maintained at high levels compared to control treatments. This increase caused tumors to subside without recurrence. We examined the immune cells mediating S.t-ΔpGlux/pT-ClyA-induced tumor suppression and examined the major cell types producing IL-1β. We found that macrophages and dendritic cells were the primary IL-1β producers. Inhibition of IL-1β in mice treated with S.t-ΔpGlux/pT-ClyA using an IL-1β antibody caused tumor growth to resume. This suggests that IL-1β plays an important role in the treatment of cancer by S.t-ΔpGlux/pT-ClyA. We found that in St-ΔpGlux/pT-ClyA-treated tumors, expression of molecules involved in signaling pathways, such as NLRP3, ASC, Caspase1, TLR4, MyD88, NF-kB and IL-1β, were upregulated, while in ΔppGpp S. typhimurium treated animals, TLR4, MyD88, NF-kB and IL-1β were upregulated with NLRP3, ASC, and Caspase1 being rarely expressed or not expressed at all. Using S.t-ΔpGlux/pT-ClyA may simultaneously activate TLR4 and NLRP3 signaling pathways, which increase IL-1β expression and enhance inhibition of colon cancer growth without tumor recurrence. This study provides a novel platform for treating colon cancer.
Collapse
|
50
|
Molinaro R, Martinez JO, Zinger A, De Vita A, Storci G, Arrighetti N, De Rosa E, Hartman KA, Basu N, Taghipour N, Corbo C, Tasciotti E. Leukocyte-mimicking nanovesicles for effective doxorubicin delivery to treat breast cancer and melanoma. Biomater Sci 2019; 8:333-341. [PMID: 31714542 DOI: 10.1039/c9bm01766f] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In the last decades, several approaches were developed to design drug delivery systems to address the multiple biological barriers encountered after administration while safely delivering a payload. In this scenario, bio-inspired and bio-mimetic approaches have emerged as promising solutions to evade the mononuclear phagocytic system while simultaneously negotiating the sequential transport across the various biological barriers. Leukocytes freely circulate in the bloodstream and selectively target the inflamed vasculature in response to injury, infection, and cancer. Recently we have shown the use of biomimetic nanovesicles, called leukosomes, which combine both the physical and biological properties of liposomes and leukocytes, respectively, to selectively deliver drugs to the inflamed vasculature. Here we report the use of leukosomes to target and deliver doxorubicin, a model chemotherapeutic, to tumors in syngeneic murine models of breast cancer and melanoma. Exploiting the inflammatory pathway responsible for recruiting immune cells to the site of injury, leukosomes exhibited increased targeting of cancer vasculature and stroma. Furthermore, delivery of doxorubicin with leukosomes enabled significant tumor growth inhibition compared with free doxorubicin in both breast and melanoma tumors. This study demonstrates the promise of using biomimetic nanovesicles for effective cancer management in solid tumors.
Collapse
Affiliation(s)
- Roberto Molinaro
- Regenerative Medicine Program, Houston Methodist Research Institute, Houston, TX, USA and School of Pharmacy, Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Jonathan O Martinez
- Regenerative Medicine Program, Houston Methodist Research Institute, Houston, TX, USA
| | - Assaf Zinger
- Regenerative Medicine Program, Houston Methodist Research Institute, Houston, TX, USA
| | - Alessandro De Vita
- Regenerative Medicine Program, Houston Methodist Research Institute, Houston, TX, USA and Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Gianluca Storci
- Regenerative Medicine Program, Houston Methodist Research Institute, Houston, TX, USA and Departmentof Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Noemi Arrighetti
- Regenerative Medicine Program, Houston Methodist Research Institute, Houston, TX, USA and Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Enrica De Rosa
- Regenerative Medicine Program, Houston Methodist Research Institute, Houston, TX, USA
| | - Kelly A Hartman
- Regenerative Medicine Program, Houston Methodist Research Institute, Houston, TX, USA
| | - Nupur Basu
- Regenerative Medicine Program, Houston Methodist Research Institute, Houston, TX, USA
| | - Nima Taghipour
- Regenerative Medicine Program, Houston Methodist Research Institute, Houston, TX, USA
| | - Claudia Corbo
- Regenerative Medicine Program, Houston Methodist Research Institute, Houston, TX, USA and School of Medicine and Surgery, Nanomedicine Center NANOMIB, University of Milano-Bicocca, Milano, Italy.
| | - Ennio Tasciotti
- Regenerative Medicine Program, Houston Methodist Research Institute, Houston, TX, USA and Houston Methodist Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, USA.
| |
Collapse
|