1
|
Di Marco T, Mazzoni M, Greco A, Cassinelli G. Non-oncogene dependencies: Novel opportunities for cancer therapy. Biochem Pharmacol 2024; 228:116254. [PMID: 38704100 DOI: 10.1016/j.bcp.2024.116254] [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: 02/07/2024] [Revised: 04/22/2024] [Accepted: 04/30/2024] [Indexed: 05/06/2024]
Abstract
Targeting oncogene addictions have changed the history of subsets of malignancies and continues to represent an excellent therapeutic opportunity. Nonetheless, alternative strategies are required to treat malignancies driven by undruggable oncogenes or loss of tumor suppressor genes and to overcome drug resistance also occurring in cancers addicted to actionable drivers. The discovery of non-oncogene addiction (NOA) uncovered novel therapeutically exploitable "Achilles' heels". NOA refers to genes/pathways not oncogenic per sé but essential for the tumor cell growth/survival while dispensable for normal cells. The clinical success of several classes of conventional and molecular targeted agents can be ascribed to their impact on both tumor cell-associated intrinsic as well as microenvironment-related extrinsic NOA. The integration of genetic, computational and pharmacological high-throughput approaches led to the identification of an expanded repertoire of synthetic lethality interactions implicating NOA targets. Only a few of them have been translated into the clinics as most NOA vulnerabilities are not easily druggable or appealing targets. Nonetheless, their identification has provided in-depth knowledge of tumor pathobiology and suggested novel therapeutic opportunities. Here, we summarize conceptual framework of intrinsic and extrinsic NOA providing exploitable vulnerabilities. Conventional and emerging methodological approaches used to disclose NOA dependencies are reported together with their limits. We illustrate NOA paradigmatic and peculiar examples and outline the functional/mechanistic aspects, potential druggability and translational interest. Finally, we comment on difficulties in exploiting the NOA-generated knowledge to develop novel therapeutic approaches to be translated into the clinics and to fully harness the potential of clinically available drugs.
Collapse
Affiliation(s)
- Tiziana Di Marco
- Integrated Biology of Rare Tumors Unit, Experimental Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy
| | - Mara Mazzoni
- Integrated Biology of Rare Tumors Unit, Experimental Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy
| | - Angela Greco
- Integrated Biology of Rare Tumors Unit, Experimental Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy
| | - Giuliana Cassinelli
- Molecular Pharmacology Unit, Experimental Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy.
| |
Collapse
|
2
|
Heo JI, Ryu J. Natural Products in the Treatment of Retinopathy of Prematurity: Exploring Therapeutic Potentials. Int J Mol Sci 2024; 25:8461. [PMID: 39126030 PMCID: PMC11313229 DOI: 10.3390/ijms25158461] [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: 05/26/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024] Open
Abstract
Retinopathy of prematurity (ROP) is a vascular disorder affecting the retinas of preterm infants. This condition arises when preterm infants in incubators are exposed to high oxygen levels, leading to oxidative stress, inflammatory responses, and a downregulation of vascular endothelial growth factors, which causes the loss of retinal microvascular capillaries. Upon returning to room air, the upregulation of vascular growth factors results in abnormal vascular growth of retinal endothelial cells. Without appropriate intervention, ROP can progress to blindness. The prevalence of ROP has risen, making it a significant cause of childhood blindness. Current treatments, such as laser therapy and various pharmacologic approaches, are limited by their potential for severe adverse effects. Therefore, a deeper understanding of ROP's pathophysiology and the development of innovative treatments are imperative. Natural products from plants, fungi, bacteria, and marine organisms have shown promise in treating various diseases and have gained attention in ROP research due to their minimal side effects and wide-ranging beneficial properties. This review discusses the roles and mechanisms of natural products that hold potential as therapeutic agents in ROP management.
Collapse
Affiliation(s)
| | - Juhee Ryu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Republic of Korea;
| |
Collapse
|
3
|
Liu J, Du H, Huang L, Xie W, Liu K, Zhang X, Chen S, Zhang Y, Li D, Pan H. AI-Powered Microfluidics: Shaping the Future of Phenotypic Drug Discovery. ACS APPLIED MATERIALS & INTERFACES 2024; 16:38832-38851. [PMID: 39016521 DOI: 10.1021/acsami.4c07665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Phenotypic drug discovery (PDD), which involves harnessing biological systems directly to uncover effective drugs, has undergone a resurgence in recent years. The rapid advancement of artificial intelligence (AI) over the past few years presents numerous opportunities for augmenting phenotypic drug screening on microfluidic platforms, leveraging its predictive capabilities, data analysis, efficient data processing, etc. Microfluidics coupled with AI is poised to revolutionize the landscape of phenotypic drug discovery. By integrating advanced microfluidic platforms with AI algorithms, researchers can rapidly screen large libraries of compounds, identify novel drug candidates, and elucidate complex biological pathways with unprecedented speed and efficiency. This review provides an overview of recent advances and challenges in AI-based microfluidics and their applications in drug discovery. We discuss the synergistic combination of microfluidic systems for high-throughput screening and AI-driven analysis for phenotype characterization, drug-target interactions, and predictive modeling. In addition, we highlight the potential of AI-powered microfluidics to achieve an automated drug screening system. Overall, AI-powered microfluidics represents a promising approach to shaping the future of phenotypic drug discovery by enabling rapid, cost-effective, and accurate identification of therapeutically relevant compounds.
Collapse
Affiliation(s)
- Junchi Liu
- Department of Anesthesiology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun 130012, China
| | - Hanze Du
- Department of Endocrinology, Key Laboratory of Endocrinology of National Health Commission, Translation Medicine Centre, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Lei Huang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, 1500 Qinghua Road, Changchun 130012, China
| | - Wangni Xie
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, 1500 Qinghua Road, Changchun 130012, China
| | - Kexuan Liu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, 1500 Qinghua Road, Changchun 130012, China
| | - Xue Zhang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, 1500 Qinghua Road, Changchun 130012, China
| | - Shi Chen
- Department of Endocrinology, Key Laboratory of Endocrinology of National Health Commission, Translation Medicine Centre, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Yuan Zhang
- Department of Anesthesiology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun 130012, China
| | - Daowei Li
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, 1500 Qinghua Road, Changchun 130012, China
| | - Hui Pan
- Department of Endocrinology, Key Laboratory of Endocrinology of National Health Commission, Translation Medicine Centre, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
4
|
Ma P, Yu H, Zhu M, Liu L, Cheng L, Han Z, Jin W. NCAPD2 promotes the malignant progression of oral squamous cell carcinoma via the Wnt/β-catenin pathway. Cell Cycle 2024; 23:588-601. [PMID: 38743408 PMCID: PMC11135826 DOI: 10.1080/15384101.2024.2348918] [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: 12/10/2021] [Accepted: 04/24/2024] [Indexed: 05/16/2024] Open
Abstract
Oral squamous cell carcinoma (OSCC) is the most common type of oral cancer, with a poor prognosis, yet the underlying mechanism needs further exploration. Non-SMC condensin I complex subunit D2 (NCAPD2) is a widely expressed protein in OSCC, but its role in tumor development is unclear. This study aimed to explore NCAPD2 expression and its biological function in OSCC. NCAPD2 expression in OSCC cell lines and tissue specimens was analyzed using quantitative polymerase chain reaction, western blotting, and immunohistochemistry. Cancer cell growth was evaluated using cell proliferation, 5-Ethynyl-2'-deoxyuridine (EdU) staining, and colony formation assays. Cell migration was evaluated using wound healing and Transwell assays. Apoptosis was detected using flow cytometry. The influence of NCAPD2 on tumor growth in vivo was evaluated in a mouse xenograft model. NCAPD2 expression was significantly higher in OSCC than that in normal oral tissue. In vitro, the knockdown of NCAPD2 inhibited OSCC cell proliferation and promoted apoptosis. NCAPD2 depletion also significantly inhibited the migration of OSCC cells. Moreover, NCAPD2 overexpression induced inverse effects on OSCC cell phenotypes. In vivo, we demonstrated that downregulating NCAPD2 could inhibit the tumorigenicity of OSCC cells. Mechanically, OSCC regulation by NCAPD2 involved the Wnt/β-catenin signaling pathway. These results suggest NCAPD2 as a novel oncogene with an important role in OSCC development and a candidate therapeutic target for OSCC.
Collapse
Affiliation(s)
- Ping Ma
- Department of Beijing Stomatological Hospital, Capital Medical University, Beijing, China
- Department of Stomatology, The Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia, China
| | - Huajiao Yu
- Department of Beijing Stomatological Hospital, Capital Medical University, Beijing, China
- Department of Stomatology, The Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia, China
| | - Mingda Zhu
- Department of Breast Disease, Henan Breast Cancer Center, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Li Liu
- Department of Stomatology, The Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia, China
| | - Luyao Cheng
- Department of Stomatology, The Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia, China
| | - Zhengxue Han
- Department of Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| | - Wulong Jin
- Department of Stomatology, The Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia, China
| |
Collapse
|
5
|
Wang Y, Yin N, Yang R, Faiola F. Pollution effects on retinal health: A review on current methodologies and findings. Toxicol Ind Health 2023; 39:336-344. [PMID: 37160417 DOI: 10.1177/07482337231174072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
In our daily life, we are exposed to numerous industrial chemicals that may be harmful to the retina, which is a delicate and sensitive part of our eyes. This could lead to irreversible changes and cause retinal diseases or blindness. Current retinal environmental health studies primarily utilize animal models, isolated mammalian retinas, animal- or human-derived retinal cells, and retinal organoids, to address both pre- and postnatal exposure. However, as there is limited toxicological information available for specific populations, human induced pluripotent stem cell (hiPSC)-induced models could be effective tools to supplement such data. In order to obtain more comprehensive and reliable toxicological information, we need more appropriate models, novel evaluation methods, and computational technologies to develop portable equipment. This review mainly focused on current toxicology models with particular emphasis on retinal organoids, and it looks forward to future models, analytical methods, and equipment that can efficiently and accurately evaluate retinal toxicity.
Collapse
Affiliation(s)
- Yue Wang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, China
| | - Nuoya Yin
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, China
| | - Renjun Yang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, China
| | - Francesco Faiola
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
6
|
Xu M, Cui Q, Su W, Zhang D, Pan J, Liu X, Pang Z, Zhu Q. High-content screening of active components of Traditional Chinese Medicine inhibiting TGF-β-induced cell EMT. Heliyon 2022; 8:e10238. [PMID: 36042745 PMCID: PMC9420491 DOI: 10.1016/j.heliyon.2022.e10238] [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: 02/25/2022] [Revised: 05/13/2022] [Accepted: 08/05/2022] [Indexed: 11/30/2022] Open
Abstract
The epithelial mesenchymal transition (EMT) has roles in metastasis and invasion during fibrotic diseases and cancer progression. Some Traditional Chinese Medicines (TCMs) have shown inhibitory effects with respect to the EMT. The current study attempted to establish a multiparametric high-content method to screen for active monomeric compounds in TCM with the ability to target cellular EMT by assessing phenotypic changes. A total of 306 monomeric compounds from the MedChemExpress (MCE) compound library were screened by the high-content screening (HCS) system and 5 compounds with anti-EMT activity, including camptothecin (CPT), dimethyl curcumin (DMC), artesunate (ART), sinapine (SNP) and berberine (BER) were identified. To confirm anti-EMT activity, expression of EMT markers was assessed by qRT-PCR and Western blotting, and cell adhesion and migration measured by cell function assays. The results revealed that CPT, DMC, ART, SNP and BER inhibited transforming growth factor-β1 (TGF-β1)-induced expression of vimentin and α-SMA, upregulated expression of E-cadherin, increased cell adhesion and reduced cell migration. In summary, by quantifying the cell morphological changes during TGF-β1-induced EMT through multi-parametric analysis, TCM compounds with anti-EMT activity were successfully screened using the HCS system, a faster and more economical approach than conventional methods.
Collapse
Affiliation(s)
- Mengzhen Xu
- College of Pharmaceutical Science, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Qinghua Cui
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Wen Su
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Dan Zhang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Jiaxu Pan
- College of Pharmaceutical Science, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Xiangqi Liu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Zheng Pang
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Qingjun Zhu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.,Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| |
Collapse
|
7
|
yingBai Y, meiCheng Y, Wang W, Yang L, Yang Y. In vivo and in vitro studies of Alloimperatorin induced autophagy in cervical cancer cells via reactive oxygen species pathway. Bioengineered 2022; 13:14299-14314. [PMID: 36708242 PMCID: PMC9995126 DOI: 10.1080/21655979.2022.2084243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/19/2022] [Accepted: 05/25/2022] [Indexed: 01/29/2023] Open
Abstract
Alloimperatorin (Alloi) has been shown to have anti-proliferative effects in our previous studies. we aimed to investigate whether Alloimperatorin induces autophagy through the reactive oxygen species (ROS) pathway and anticancer activity in vivo. The anti-proliferative effect of Alloimperatorin was evaluated using a cell counting kit (CCK-8 kit). Apoptosis was detected using flow cytometry. Confocal microscopy, immunofluorescence, and mRFP-GFP-LC3 lentivirus transfection were used to verify autophagy. Electron microscopy detection of autophagosomes was induced by Alloimperatorin. Western blotting was used to detect autophagy proteins in HeLa and SiHa cells. A xenograft model was used to monitor the inhibitory effect of Alloimperatorin on tumor growth in nude mice. The results showed that Alloimperatorin induced ROS production and inhibited the proliferation of HeLa and SiHa cells. Furthermore, Alloimperatorin increased the apoptosis rate in HeLa and SiHa cells. Confocal microscopy fluorescence indicated that Alloimperatorin increased autophagy fluorescence of HeLa and SiHa cells. mRFP-GFP-LC3 lentivirus transfection and electron microscopy demonstrated that Alloimperatorin increased autophagy in HeLa and SiHa cells. Western blotting showed that Alloimperatorin induced the expression of autophagy proteins in HeLa and SiHa cells. However, N-acetylcysteine reversed the autophagy. These results demonstrate that Alloimperatorin can induce autophagy in HeLa and SiHa cells through the ROS pathway. In vivo xenograft experiments showed that Alloimperatorin could inhibit tumor growth in nude mice. Alloimperatorin is expected to be an effective new drug for cervical cancer treatment.Abbreviations: ROS, reactive oxygen species; Alloi, Alloimperatorin; CCK-8, Cell Counting Kit-8; NAC, N-acetyl-L-cysteine; DCFH-DA, 2,7-dichlorodihydrofluorescein diacetate; OD, optical density; PBS, phosphate buffer solution; BCA, bicinchoninic acid; DAPI, 4,6-diamidino-2-phenylindole; DMSO, dimethyl sulfoxide.
Collapse
Affiliation(s)
- Ying yingBai
- The First Clinical Medical College of Lanzhou University, Lanzhou, PR China
| | - Yue meiCheng
- The First Clinical Medical College of Lanzhou University, Lanzhou, PR China
| | - Wenhua Wang
- The First Clinical Medical College of Lanzhou University, Lanzhou, PR China
| | - Lijuan Yang
- The First Clinical Medical College of Lanzhou University, Lanzhou, PR China
| | - Yongxiu Yang
- The First Clinical Medical College of Lanzhou University, Lanzhou, PR China
- Department of Obstetrics and Gynecology, First Hospital of Lanzhou University, Lanzhou, PR China
| |
Collapse
|
8
|
Han L, Yuan Y, Chen X, Huang J, Wang G, Zhou C, Dong J, Zhang N, Zhang Y, Yin H, Jiang Y. A Candidate Drug Screen Strategy: The Discovery of Oroxylin A in Scutellariae Radix Against Sepsis via the Correlation Analysis Between Plant Metabolomics and Pharmacodynamics. Front Pharmacol 2022; 13:861105. [PMID: 35662699 PMCID: PMC9160923 DOI: 10.3389/fphar.2022.861105] [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: 01/24/2022] [Accepted: 03/25/2022] [Indexed: 11/13/2022] Open
Abstract
Sepsis is an acute systemic infectious disease with high mortality, which urgently needs more effective treatment. Scutellariae radix (SR), a commonly used traditional Chinese medicine (TCM) for clearing heat and detoxification, contains rich natural products possessing anti-inflammatory activity. In previous studies, it was found that the anti-inflammatory activities of SR extracts from different ecological conditions varied wildly. Based on this, in the present study, a screening strategy of antisepsis active components from SR based on correlation analysis between plant metabolomics and pharmacodynamics was established, and the mechanism was explored. First of all, a mass spectrum database of SR (above 240 components) was established to lay the foundation for the identification of plant metabolomics by liquid chromatography tandem mass spectrometry (LC-MS/MS). Through the correlation analysis between plant metabolomics and anti-inflammatory activity of SR from different ecology regions, 10 potential components with high correlation coefficients were preliminarily screened out. After the evaluation of anti-inflammatory activity and toxicity at the cellular level, the pharmacodynamic evaluation in vivo found that oroxylin A had the potentiality of antisepsis both in LPS- and CLP-induced endotoxemia mice. Network pharmacology and Western blot (WB) results indicated that oroxylin A significantly inhibited the toll-like receptor 4/nuclear factor-kappa B (TLR4/NF-κB) signaling pathway, which was further confirmed by secreted embryonic alkaline phosphatase (SEAP) assay. Moreover, the molecular docking analysis indicated that oroxylin A might competitively inhibit LPS binding to myeloid differentiation 2 (MD-2) to block the activation of TLR4. The study provided a feasible research strategy for the screening and discovery of antisepsis candidate drugs from TCM.
Collapse
Affiliation(s)
- Lingyu Han
- School of Pharmaceutical Sciences, Institute for Chinese Materia Medica, Tsinghua University, Beijing, China
| | - Yue Yuan
- School of Pharmaceutical Sciences, Institute for Chinese Materia Medica, Tsinghua University, Beijing, China
| | - Xinyi Chen
- School of Pharmaceutical Sciences, Institute for Chinese Materia Medica, Tsinghua University, Beijing, China
| | - Jian Huang
- Key Laboratory of Bioorganic Phosphorous Chemistry and Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
- Department of Molecular Biology, Princeton University, Princeton, NJ, United States
| | - Guan Wang
- Beijing Huisheng Biotechnology Co., Ltd., Beijing, China
| | - Chao Zhou
- Waters Technologies (Shanghai) Ltd., Beijing, China
| | - Jianjian Dong
- School of Pharmaceutical Sciences, Institute for Chinese Materia Medica, Tsinghua University, Beijing, China
| | - Na Zhang
- School of Pharmaceutical Sciences, Institute for Chinese Materia Medica, Tsinghua University, Beijing, China
| | - Yuxin Zhang
- Key Laboratory of Bioorganic Phosphorous Chemistry and Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Hang Yin
- School of Pharmaceutical Sciences, Institute for Chinese Materia Medica, Tsinghua University, Beijing, China
- Key Laboratory of Bioorganic Phosphorous Chemistry and Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Yunyao Jiang
- School of Pharmaceutical Sciences, Institute for Chinese Materia Medica, Tsinghua University, Beijing, China
| |
Collapse
|
9
|
The Antitumoral/Antimetastatic Action of the Flavonoid Brachydin A in Metastatic Prostate Tumor Spheroids In Vitro Is Mediated by (Parthanatos) PARP-Related Cell Death. Pharmaceutics 2022; 14:pharmaceutics14050963. [PMID: 35631550 PMCID: PMC9147598 DOI: 10.3390/pharmaceutics14050963] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/23/2022] [Accepted: 04/26/2022] [Indexed: 12/13/2022] Open
Abstract
Metastatic prostate cancer (mPCa) is resistant to several chemotherapeutic agents. Brachydin A (BrA), a glycosylated flavonoid extracted from Fridericia platyphylla, displays a remarkable antitumoral effect against in vitro mPCa cells cultured as bidimensional (2D) monolayers. Considering that three-dimensional (3D) cell cultures provide a more accurate response to chemotherapeutic agents, this study investigated the antiproliferative/antimetastatic effects of BrA and the molecular mechanisms underlying its action in mPCa spheroids (DU145) in vitro. BrA at 60–100 μM was cytotoxic, altered spheroid morphology/volume, and suppressed cell migration and tumor invasiveness. High-content analysis revealed that BrA (60–100 µM) reduced mitochondrial membrane potential and increased apoptosis and necrosis markers, indicating that it triggered cell death mechanisms. Molecular analysis showed that (i) 24-h treatment with BrA (80–100 µM) increased the protein levels of DNA disruption markers (cleaved-PARP and p-γ-H2AX) as well as decreased the protein levels of anti/pro-apoptotic (BCL-2, BAD, and RIP3K) and cell survival markers (p-AKT1 and p-44/42 MAPK); (ii) 72-h treatment with BrA increased the protein levels of effector caspases (CASP3, CASP7, and CASP8) and inflammation markers (NF-kB and TNF-α). Altogether, our results suggest that PARP-mediated cell death (parthanatos) is a potential mechanism of action. In conclusion, BrA confirms its potential as a candidate drug for preclinical studies against mPCa.
Collapse
|
10
|
Decarli MC, Mizukami A, Azoubel RA, Neto PI, Mota C, Moraes ÂM, Silva JVL, Moroni L. Static systems to obtain 3D spheroid cell models: a cost analysis comparing the implementation of four types of microwell array inserts. Biochem Eng J 2022. [DOI: 10.1016/j.bej.2022.108414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
11
|
Decarli MC, de Castro MV, Nogueira JA, Nagahara MHT, Westin CB, de Oliveira ALR, Silva JVL, Moroni L, Mota C, Moraes ÂM. Development of a device useful to reproducibly produce large quantities of viable and uniform stem cell spheroids with controlled diameters. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2022; 135:112685. [DOI: 10.1016/j.msec.2022.112685] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 12/20/2021] [Accepted: 01/22/2022] [Indexed: 01/08/2023]
|
12
|
Bonnekoh H, Vera C, Abad‐Perez A, Radetzki S, Neuenschwander M, Specker E, Mahnke NA, Frischbutter S, Latz E, Nazaré M, Kries JV, Maurer M, Scheffel J, Krause K. Topical inflammasome inhibition with disulfiram prevents irritant contact dermatitis. Clin Transl Allergy 2021; 11:e12045. [PMID: 34322217 PMCID: PMC8297992 DOI: 10.1002/clt2.12045] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 07/01/2021] [Accepted: 07/08/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND The pathogenesis of contact dermatitis, a common inflammatory skin disease with limited treatment options, is held to be driven by inflammasome activation induced by allergens and irritants. We here aim to identify inflammasome-targeting treatment strategies for irritant contact dermatitis. METHODS A high content screen with 41,184 small molecules was performed using fluorescent Apoptosis associated speck-like protein containing a CARD (ASC) speck formation as a readout for inflammasome activation. Hit compounds were validated for inhibition of interleukin (IL)-1β secretion. Of these, the approved thiuramdisulfide derivative disulfiram was selected and tested in a patch test model of irritant contact dermatitis in 25 healthy volunteers. Topical application of disulfiram, mometasone or vehicle was followed by application of sodiumdodecylsulfate (SDS) for 24 h each. Eczema induction was quantified by mexameter and laser speckle imaging. Corneocyte sampling of lesional skin was performed to assess inflammasome-mediated cytokines IL-1β and IL-18. RESULTS Disulfiram induced a dose-dependent inhibition of ASC speck formation and IL-1β release in cellular assays in vitro. In vivo, treatment with disulfiram, but not with vehicle and less mometasone, inhibited SDS-induced eczema. This was demonstrated by significantly lower erythema and total perfusion values assessed by mexameter and laser speckle imaging for disulfiram compared to vehicle (p < 0.001) and/or mometasone (p < 0.001). Also, corneocyte IL-18 levels were significantly reduced after application of disulfiram compared to vehicle (p < 0.001). CONCLUSION We show that disulfiram is a dose-dependent inhibitor of inflammasome pathway activation in vitro and inhibitor of SDS-induced eczema in vivo. Topical application of disulfiram represents a potential treatment option for irritant contact dermatitis.
Collapse
Affiliation(s)
- Hanna Bonnekoh
- Dermatological Allergology, Allergie‐Centrum‐Charité, Department of Dermatology, Venereology and AllergologyCharité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
- Autoinflammation Reference Center Charité (ARC2)Charité – Universitätsmedizin BerlinBerlinGermany
| | - Carolina Vera
- Dermatological Allergology, Allergie‐Centrum‐Charité, Department of Dermatology, Venereology and AllergologyCharité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
- Autoinflammation Reference Center Charité (ARC2)Charité – Universitätsmedizin BerlinBerlinGermany
| | - Angela Abad‐Perez
- Dermatological Allergology, Allergie‐Centrum‐Charité, Department of Dermatology, Venereology and AllergologyCharité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
- Autoinflammation Reference Center Charité (ARC2)Charité – Universitätsmedizin BerlinBerlinGermany
| | - Silke Radetzki
- Department of Chemical BiologyLeibniz‐Forschungsinstitut für Molekulare Pharmakologie (FMP)BerlinGermany
| | - Martin Neuenschwander
- Department of Chemical BiologyLeibniz‐Forschungsinstitut für Molekulare Pharmakologie (FMP)BerlinGermany
| | - Edgar Specker
- Department of Chemical BiologyLeibniz‐Forschungsinstitut für Molekulare Pharmakologie (FMP)BerlinGermany
| | - Niklas Amadeus Mahnke
- Dermatological Allergology, Allergie‐Centrum‐Charité, Department of Dermatology, Venereology and AllergologyCharité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
- Autoinflammation Reference Center Charité (ARC2)Charité – Universitätsmedizin BerlinBerlinGermany
| | - Stefan Frischbutter
- Dermatological Allergology, Allergie‐Centrum‐Charité, Department of Dermatology, Venereology and AllergologyCharité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
- Autoinflammation Reference Center Charité (ARC2)Charité – Universitätsmedizin BerlinBerlinGermany
| | - Eicke Latz
- Institute of Innate ImmunityUniversity of BonnBonnGermany
- German Center of Neurodegenerative Diseases (DZNE)University of BonnBonnGermany
| | - Marc Nazaré
- Department of Chemical BiologyLeibniz‐Forschungsinstitut für Molekulare Pharmakologie (FMP)BerlinGermany
| | - Jens v. Kries
- Department of Chemical BiologyLeibniz‐Forschungsinstitut für Molekulare Pharmakologie (FMP)BerlinGermany
| | - Marcus Maurer
- Dermatological Allergology, Allergie‐Centrum‐Charité, Department of Dermatology, Venereology and AllergologyCharité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
- Autoinflammation Reference Center Charité (ARC2)Charité – Universitätsmedizin BerlinBerlinGermany
| | - Jörg Scheffel
- Dermatological Allergology, Allergie‐Centrum‐Charité, Department of Dermatology, Venereology and AllergologyCharité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
- Autoinflammation Reference Center Charité (ARC2)Charité – Universitätsmedizin BerlinBerlinGermany
| | - Karoline Krause
- Dermatological Allergology, Allergie‐Centrum‐Charité, Department of Dermatology, Venereology and AllergologyCharité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
- Autoinflammation Reference Center Charité (ARC2)Charité – Universitätsmedizin BerlinBerlinGermany
| |
Collapse
|
13
|
Smith EJ, Farshim PP, Flomen R, Jones ST, McAteer SJ, Deverman BE, Gradinaru V, Bates GP. Use of high-content imaging to quantify transduction of AAV-PHP viruses in the brain following systemic delivery. Brain Commun 2021; 3:fcab105. [PMID: 34131644 PMCID: PMC8200048 DOI: 10.1093/braincomms/fcab105] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 03/19/2021] [Accepted: 04/08/2021] [Indexed: 12/26/2022] Open
Abstract
The engineering of the AAV-PHP capsids was an important development for CNS research and the modulation of gene expression in the brain. They cross the blood brain barrier and transduce brain cells after intravenous systemic delivery, a property dependent on the genotype of Ly6a, the AAV-PHP capsid receptor. It is important to determine the transduction efficiency of a given viral preparation, as well as the comparative tropism for different brain cells; however, manual estimation of adeno-associated viral transduction efficiencies can be biased and time consuming. Therefore, we have used the Opera Phenix high-content screening system, equipped with the Harmony processing and analysis software, to reduce bias and develop an automated approach to determining transduction efficiency in the mouse brain. We used R Studio and 'gatepoints' to segment the data captured from coronal brain sections into brain regions of interest. C57BL/6J and CBA/Ca mice were injected with an AAV-PHP.B virus containing a green fluorescent protein reporter with a nuclear localization signal. Coronal sections at 600 μm intervals throughout the entire brain were stained with Hoechst dye, combined with immunofluorescence to NeuN and green fluorescent protein to identify all cell nuclei, neurons and transduced cells, respectively. Automated data analysis was applied to give an estimate of neuronal percentages and transduction efficiencies throughout the entire brain as well as for the cortex, striatum and hippocampus. The data from each coronal section from a given mouse were highly comparable. The percentage of neurons in the C57BL/6J and CBA/Ca brains was approximately 40% and this was higher in the cortex than striatum and hippocampus. The systemic injection of AAV-PHP.B resulted in similar transduction rates across the entire brain for C57BL/6J mice. Approximately 10-15% of all cells were transduced, with neuronal transduction efficiencies ranging from 5% to 15%, estimates that were similar across brain regions, and were in contrast to the much more localized transduction efficiencies achieved through intracerebral injection. We confirmed that the delivery of the AAV-PHP.B viruses to the brain from the vasculature resulted in widespread transduction. Our methodology allows the rapid comparison of transduction rates between brain regions producing comparable data to more time-consuming approaches. The methodology developed here can be applied to the automated quantification of any parameter of interest that can be captured as a fluorescent signal.
Collapse
Affiliation(s)
- Edward J Smith
- Huntington’s Disease Centre, Department of Neurodegenerative Disease, UK Dementia Research Institute at UCL, Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Pamela P Farshim
- Huntington’s Disease Centre, Department of Neurodegenerative Disease, UK Dementia Research Institute at UCL, Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Rachel Flomen
- Huntington’s Disease Centre, Department of Neurodegenerative Disease, UK Dementia Research Institute at UCL, Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Samuel T Jones
- Huntington’s Disease Centre, Department of Neurodegenerative Disease, UK Dementia Research Institute at UCL, Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Sean J McAteer
- Huntington’s Disease Centre, Department of Neurodegenerative Disease, UK Dementia Research Institute at UCL, Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Benjamin E Deverman
- The Stanley Center for Psychiatric Research at the Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Viviana Gradinaru
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91101, USA
| | - Gillian P Bates
- Huntington’s Disease Centre, Department of Neurodegenerative Disease, UK Dementia Research Institute at UCL, Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| |
Collapse
|
14
|
Nuthan BR, Rakshith D, Marulasiddaswamy KM, Rao HCY, Ramesha KP, Mohana NC, Siddappa S, Darshan D, Kumara KKS, Satish S. Application of Optimized and Validated Agar Overlay TLC-Bioautography Assay for Detecting the Antimicrobial Metabolites of Pharmaceutical Interest. J Chromatogr Sci 2021; 58:737-746. [PMID: 32766714 DOI: 10.1093/chromsci/bmaa045] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Indexed: 02/06/2023]
Abstract
The agar overlay TLC-bioautography is one of the crucial methods for simultaneous in situ detection and separation of antimicrobial metabolites of pharmaceutical interest. The main focus of this research relies on the dereplication of an antimicrobial metabolite coriloxin derived from mycoendophytic Xylaria sp. NBRTSB-20 with a validation of agar overlay TLC-bioautography technique. This polyketide metabolite coriloxin was purified by column chromatography, and its purity was assessed by HPLC, UPLC-ESI-QTOF-MS, FT-IR and NMR spectral analysis. The antimicrobial capability of ethyl acetate extract and the purified compound coriloxin was determined by disc diffusion, minimal inhibitory concentration and agar overlay TLC-bioautography assay. The visible LOD of coriloxin antimicrobial activity was found at 10 μg for Escherichia coli and 20 μg for both Staphylococcus aureus and Fusarium oxysporum. Inter- and intra-day precision was determined as the relative standard deviation is less than 6.56%, which proved that this method was precise. The accuracy was expressed as recovery, and the values were found ranging from 91.18 to 108.73% with RSD values 0.94-2.30%, respectively. The overall findings of this investigation suggest that agar overlay TLC-bioautography assay is a suitable and acceptable method for the in situ determination of antimicrobial pharmaceuticals.
Collapse
Affiliation(s)
| | - Devaraju Rakshith
- Department of Studies in Microbiology, University of Mysore, Manasagangotri, Mysuru 570 006, India
| | | | - H C Yashavantha Rao
- Department of Biochemistry, Indian Institute of Science, Bengaluru 560 012, Karnataka, India
| | | | | | - Shiva Siddappa
- Department of Studies in Biochemistry, University of Mysore, Manasagangotri, Mysuru 570 006, India
| | - Doreraj Darshan
- Plant Cell Biotechnology, CSIR-Central Food Technological Research Institute, Mysuru 570 005, India
| | | | - Sreedharamurthy Satish
- Department of Studies in Microbiology, University of Mysore, Manasagangotri, Mysuru 570 006, India
| |
Collapse
|
15
|
Decarli MC, do Amaral RLF, Dos Santos DP, Tofani LB, Katayama E, Rezende RA, Silva JVLD, Swiech K, Suazo CAT, Mota C, Moroni L, Moraes ÂM. Cell spheroids as a versatile research platform: formation mechanisms, high throughput production, characterization and applications. Biofabrication 2021; 13. [PMID: 33592595 DOI: 10.1088/1758-5090/abe6f2] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 02/16/2021] [Indexed: 11/12/2022]
Abstract
Three-dimensional cell culture has tremendous advantages to closely mimic the in vivo architecture and microenvironment of healthy tissue and organs, as well as of solid tumors. Spheroids are currently the most attractive 3D model to produce uniform reproducible cell structures as well as a potential basis for engineering large tissues and complex organs. In this review we discuss, from an engineering perspective, processes to obtain uniform 3D cell spheroids, comparing dynamic and static cultures and considering aspects such as mass transfer and shear stress. In addition, computational and mathematical modelling of complex cell spheroid systems are discussed. The non-cell-adhesive hydrogel-based method and dynamic cell culture in bioreactors are focused in detail and the myriad of developed spheroid characterization techniques is presented. The main bottlenecks and weaknesses are discussed, especially regarding the analysis of morphological parameters, cell quantification and viability, gene expression profiles, metabolic behavior and high-content analysis. Finally, a vast set of applications of spheroids as tools for in vitro study model systems is examined, including drug screening, tissue formation, pathologies development, tissue engineering and biofabrication, 3D bioprinting and microfluidics, together with their use in high-throughput platforms.
Collapse
Affiliation(s)
- Monize Caiado Decarli
- School of Chemical Engineering/Department of Engineering of Materials and of Bioprocesses, University of Campinas, Av. Albert Einstein, 500 - Bloco A - Cidade Universitária Zeferino Vaz, Cidade Universitária Zeferino Vaz, Campinas, SP, 13083-852, BRAZIL
| | - Robson Luis Ferraz do Amaral
- School of Pharmaceutical Sciences of Ribeirão Preto/Department of Pharmaceutical Sciences, University of São Paulo, Avenida do Café, no number, Ribeirão Preto, SP, 14040-903, BRAZIL
| | - Diogo Peres Dos Santos
- Departament of Chemical Engineering, Federal University of São Carlos, Rod. Washington Luiz (SP-310), km 235, São Carlos, SP, 13565-905, BRAZIL
| | - Larissa Bueno Tofani
- School of Pharmaceutical Sciences of Ribeirão Preto/Department of Pharmaceutical Sciences, University of São Paulo, Avenida do Café, no number, Ribeirão Preto, SP, 14040-903, BRAZIL
| | - Eric Katayama
- Departament of Chemical Engineering, Federal University of São Carlos, Rod. Washington Luiz (SP-310), km 235, São Carlos, SP, 13565-905, BRAZIL
| | - Rodrigo Alvarenga Rezende
- Centro de Tecnologia da Informacao Renato Archer, Rod. Dom Pedro I (SP-65), km 143,6 - Amarais, Campinas, SP, 13069-901, BRAZIL
| | - Jorge Vicente Lopes da Silva
- Centro de Tecnologia da Informacao Renato Archer, Rod. Dom Pedro I (SP-65), km 143,6 - Amarais, Campinas, SP, 13069-901, BRAZIL
| | - Kamilla Swiech
- University of Sao Paulo, School of Pharmaceutical Sciences of Ribeirão Preto/Department of Pharmaceutical Sciences, Ribeirao Preto, SP, 14040-903, BRAZIL
| | - Cláudio Alberto Torres Suazo
- Department of Chemical Engineering, Federal University of São Carlos, Rod. Washington Luiz (SP-310), km 235, São Carlos, SP, 13565-905, BRAZIL
| | - Carlos Mota
- Department of Complex Tissue Regeneration (CTR), University of Maastricht , Universiteitssingel, 40, office 3.541A, Maastricht, 6229 ER, NETHERLANDS
| | - Lorenzo Moroni
- Complex Tissue Regeneration, Maastricht University, Universiteitsingel, 40, Maastricht, 6229ER, NETHERLANDS
| | - Ângela Maria Moraes
- School of Chemical Engineering/Department of Engineering of Materials and of Bioprocesses, University of Campinas, Av. Albert Einstein, 500 - Bloco A - Cidade Universitária Zeferino Vaz, Campinas, SP, 13083-852, BRAZIL
| |
Collapse
|
16
|
Pringle NA, van de Venter M, Koekemoer TC. Comprehensive in vitro antidiabetic screening of Aspalathus linearis using a target-directed screening platform and cellomics. Food Funct 2021; 12:1020-1038. [PMID: 33416070 DOI: 10.1039/d0fo02611e] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The antidiabetic potential of Aspalathus linearis has been investigated for over a decade, however, its characterisation remains incomplete with results scattered across numerous journals making the information difficult to compare and integrate. To explore whether any potential antidiabetic mechanisms for A. linearis have been neglected and to compare the suitability of extracts of green and "fermented" A. linearis as potential antidiabetic treatment strategies, this study utilised a comprehensive in vitro antidiabetic target-directed screening platform in combination with high content screening and analysis/cellomics. The antidiabetic screening platform consisted of 20 different screening assays that incorporated 5 well-characterised antidiabetic targets i.e. the intestine, liver, skeletal muscle, adipose tissue/obesity and pancreatic β-cells. Both the green and fermented extracts of A. linearis demonstrated very broad antidiabetic mechanisms as they revealed several promising activities that could be useful in combatting insulin resistance, inflammation, oxidative stress, protein glycation and pancreatic β-cell dysfunction and death - with a strong tendency to attenuate postprandial hyperglycaemia and the subsequent metabolic dysfunction which arises as a result of poor glycaemic control. The green extract was more successful at combatting oxidative stress in INS-1 pancreatic β-cells and enhancing intracellular calcium levels in the absence of glucose. Conversely, the fermented extract demonstrated a greater ability to inhibit α-glucosidase activity as well as palmitic acid-induced free fatty acid accumulation in C3A hepatocytes and differentiated L6 myotubes, however, further studies are required to clarify the potentially toxic and pro-inflammatory nature of the fermented extract.
Collapse
Affiliation(s)
- Nadine A Pringle
- Department of Biochemistry and Microbiology, Nelson Mandela University, Port Elizabeth, South Africa.
| | - Maryna van de Venter
- Department of Biochemistry and Microbiology, Nelson Mandela University, Port Elizabeth, South Africa.
| | - Trevor C Koekemoer
- Department of Biochemistry and Microbiology, Nelson Mandela University, Port Elizabeth, South Africa.
| |
Collapse
|
17
|
He Y, Chen S, Tsoi B, Qi S, Gu B, Wang Z, Peng C, Shen J. Alpinia oxyphylla Miq. and Its Active Compound P-Coumaric Acid Promote Brain-Derived Neurotrophic Factor Signaling for Inducing Hippocampal Neurogenesis and Improving Post-cerebral Ischemic Spatial Cognitive Functions. Front Cell Dev Biol 2021; 8:577790. [PMID: 33537297 PMCID: PMC7849625 DOI: 10.3389/fcell.2020.577790] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 12/07/2020] [Indexed: 01/19/2023] Open
Abstract
Alpinia oxyphylla Miq. (AOM) is a medicinal herb for improving cognitive functions in traditional Chinese medicine for poststroke treatment, but its efficacies and underlying mechanisms remain unknown. In the present study, we tested the hypothesis that AOM could induce adult hippocampal neurogenesis and improve poststroke cognitive impairment via inducing brain-derived neurotrophic factor (BDNF) signaling pathway. In order to test the hypothesis, we performed both in vivo rat experiments using transient middle cerebral artery occlusion (MCAO) model and in vitro neural stem cell (NSC) experiments using oxygen–glucose deprivation plus reoxygenation. First, AOM treatment significantly up-regulated the expression of BDNF, tropomycin receptor kinase B (TrkB), and phosphorylated AKT (p-AKT) in the hippocampus, enhanced adult hippocampal neurogenesis, and improved the spatial learning/memory and cognitive functions in the post-MCAO ischemic rats in vivo. Next, in vitro studies confirmed p-coumaric acid (P-CA) to be the most effective compound identified from AOM extract with the properties of activating BDNF/TrkB/AKT signaling pathway and promoting NSC proliferation. Cotreatment of BDNF/TrkB-specific inhibitor ANA12 abolished the effects of P-CA on inducing BDNF/TrkB/AKT activation and the NSC proliferation. Finally, animal experiments showed that P-CA treatment enhanced the neuronal proliferation and differentiation in the hippocampus, improved spatial learning and memory functions, and reduced anxiety in the transient MCAO ischemic rats. In conclusion, P-CA is a representative compound from AOM for its bioactivities of activating BDNF/TrkB/AKT signaling pathway, promoting hippocampal neurogenesis, improving cognitive functions, and reducing anxiety in post–ischemic stroke rats.
Collapse
Affiliation(s)
- Yacong He
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Shuang Chen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Bun Tsoi
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Shuhua Qi
- Medical Technology School, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou, China
| | - Bing Gu
- Medical Technology School, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou, China
| | - Zhenxing Wang
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Cheng Peng
- Key Laboratory of Standardization of Chinese Herbal Medicines of Ministry of Education, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiangang Shen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.,Medical Technology School, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
18
|
Salikin NH, Nappi J, Majzoub ME, Egan S. Combating Parasitic Nematode Infections, Newly Discovered Antinematode Compounds from Marine Epiphytic Bacteria. Microorganisms 2020; 8:E1963. [PMID: 33322253 PMCID: PMC7764037 DOI: 10.3390/microorganisms8121963] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/08/2020] [Accepted: 12/08/2020] [Indexed: 02/06/2023] Open
Abstract
Parasitic nematode infections cause debilitating diseases and impede economic productivity. Antinematode chemotherapies are fundamental to modern medicine and are also important for industries including agriculture, aquaculture and animal health. However, the lack of suitable treatments for some diseases and the rise of nematode resistance to many available therapies necessitates the discovery and development of new drugs. Here, marine epiphytic bacteria represent a promising repository of newly discovered antinematode compounds. Epiphytic bacteria are ubiquitous on marine surfaces where they are under constant pressure of grazing by bacterivorous predators (e.g., protozoans and nematodes). Studies have shown that these bacteria have developed defense strategies to prevent grazers by producing toxic bioactive compounds. Although several active metabolites against nematodes have been identified from marine bacteria, drug discovery from marine microorganisms remains underexplored. In this review, we aim to provide further insight into the need and potential for marine epiphytic bacteria to become a new source of antinematode drugs. We discuss current and emerging strategies, including culture-independent high throughput screening and the utilization of Caenorhabditis elegans as a model target organism, which will be required to advance antinematode drug discovery and development from marine microbial sources.
Collapse
Affiliation(s)
- Nor Hawani Salikin
- Centre for Marine Science and Innovation, School of Biological, Earth and Environmental Sciences, UNSW, Sydney, NSW 2052, Australia; (N.H.S.); (J.N.); (M.E.M.)
- School of Industrial Technology, Universiti Sains Malaysia, USM, 11800 Penang, Malaysia
| | - Jadranka Nappi
- Centre for Marine Science and Innovation, School of Biological, Earth and Environmental Sciences, UNSW, Sydney, NSW 2052, Australia; (N.H.S.); (J.N.); (M.E.M.)
| | - Marwan E. Majzoub
- Centre for Marine Science and Innovation, School of Biological, Earth and Environmental Sciences, UNSW, Sydney, NSW 2052, Australia; (N.H.S.); (J.N.); (M.E.M.)
| | - Suhelen Egan
- Centre for Marine Science and Innovation, School of Biological, Earth and Environmental Sciences, UNSW, Sydney, NSW 2052, Australia; (N.H.S.); (J.N.); (M.E.M.)
| |
Collapse
|
19
|
Rossi O, Vlazaki M, Kanvatirth P, Restif O, Mastroeni P. Within-host spatiotemporal dynamic of systemic salmonellosis: Ways to track infection, reaction to vaccination and antimicrobial treatment. J Microbiol Methods 2020; 176:106008. [PMID: 32707153 DOI: 10.1016/j.mimet.2020.106008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 07/13/2020] [Accepted: 07/17/2020] [Indexed: 12/16/2022]
Abstract
During the last two decades our understanding of the complex in vivo host-pathogen interactions has increased due to technical improvements and new research tools. The rapid advancement of molecular biology, flow cytometry and microscopy techniques, combined with mathematical modelling, have empowered in-depth studies of systemic bacterial infections across scales from single molecules, to cells, to organs and systems to reach the whole organism level. By tracking subpopulations of bacteria in vivo using molecular or fluorescent tags, it has been possible to reconstruct the spread of infection within and between organs, allowing unprecedented quantification of the effects of antimicrobial treatment and vaccination. This review illustrates recent advances in the study of heterogeneous traits of the infection process and illustrate approaches to investigate the reciprocal interactions between antimicrobial treatments, bacterial growth/death as well as inter- and intra-organ spread. We also discuss how vaccines impact the in vivo behaviour of bacteria and how these findings can guide vaccine design and rational antimicrobial treatment.
Collapse
Affiliation(s)
- Omar Rossi
- University of Cambridge, Department of Veterinary Medicine, Madingley Road, CB3 0ES Cambridge, UK.
| | - Myrto Vlazaki
- University of Cambridge, Department of Veterinary Medicine, Madingley Road, CB3 0ES Cambridge, UK
| | - Panchali Kanvatirth
- University of Cambridge, Department of Veterinary Medicine, Madingley Road, CB3 0ES Cambridge, UK
| | - Olivier Restif
- University of Cambridge, Department of Veterinary Medicine, Madingley Road, CB3 0ES Cambridge, UK
| | - Pietro Mastroeni
- University of Cambridge, Department of Veterinary Medicine, Madingley Road, CB3 0ES Cambridge, UK.
| |
Collapse
|
20
|
Guo R, Liu N, Liu H, Zhang J, Zhang H, Wang Y, Baruscotti M, Zhao L, Wang Y. High content screening identifies licoisoflavone A as a bioactive compound of Tongmaiyangxin Pills to restrain cardiomyocyte hypertrophy via activating Sirt3. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 68:153171. [PMID: 32018211 DOI: 10.1016/j.phymed.2020.153171] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 12/21/2019] [Accepted: 01/09/2020] [Indexed: 06/10/2023]
Abstract
BACKGROUND Cardiac hypertrophy is a prominent feature of heart remodeling, which may eventually lead to heart failure. Tongmaiyangxin (TMYX) pills are a clinically used botanical drug for treating multiple cardiovascular diseases including chronic heart failure. The aim of the current study was to identify the bioactive compounds in Tongmaiyangxin pills that attenuate cardiomyocytes hypertrophy, and to investigate the underlying mechanism of action. METHODS AND RESULTS The anti-hypertrophy effect of TMYX was validated in isoproterenol-induced cardiac hypertrophy model in C57BL/6 mice. After TMYX treatment for 2 weeks, the heart ejection fraction and fractional shortening of the mice model was increased by approximately 20% and 15%, respectively, (p < 0.05). Besides, TMYX dose-dependently reduced the cross section area of cardiomyocytes in the angiotensin-II induced hypertrophy H9c2 model (p < 0.01). Combining high content screening and liquid chromatography mass spectrometry, four compounds with anti-cardiac hypertrophy effects were identified from TMYX, which includes emodin, licoisoflavone A, licoricone and glyasperin A. Licoisoflavone A is one of the compounds with most significant protective effect and we continued to investigate the mechanism. Primary cultures of neonatal rat cardiomyocytes were treated with a hypertrophic agonist phenylephrine (PE) in the presence or absence of licoisoflavone A. After 48 h of treatment, cells were harvested and mitochondrial acetylation was analyzed by western blotting and Image analysis. Interestingly, the results suggested that the anti-hypertrophic effects of licoisoflavone A depend on the activation of the deacetylase Sirt3 (p < 0.01). Finally, we showed that licoisoflavone A-treatment was able to decrease relative ANF and BNP levels in the hypertrophic cardiac cells (p < 0.01), but not in cells co-treated with Sirt3 inhibitors (3-TYP) (p > 0.05). CONCLUSION TMYX exerts its anti-hypertrophy effect possibly through upregulating Sirt3 expression. Four compounds were identified from TMYX which may be responsible for the anti-hypertrophy effect. Among these compounds, licoisoflavone A was demonstrated to block the hypertrophic response of cardiomyocytes, which required its positive regulation on the expression of Sirt3. These results suggested that licoisoflavone A is a potential Sirt3 activator with therapeutic effect on cardiac hypertrophy.
Collapse
MESH Headings
- Acetylation
- Angiotensin II/adverse effects
- Animals
- Cardiomegaly/chemically induced
- Cardiomegaly/drug therapy
- Cells, Cultured
- Disease Models, Animal
- Drug Evaluation, Preclinical
- Drugs, Chinese Herbal/chemistry
- Drugs, Chinese Herbal/pharmacology
- Isoflavones/pharmacology
- Isoproterenol/adverse effects
- Male
- Mice, Inbred C57BL
- Mitochondria, Heart/drug effects
- Mitochondria, Heart/metabolism
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Phenylephrine/adverse effects
- Rats
- Sirtuin 3/metabolism
Collapse
Affiliation(s)
- Rui Guo
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ningning Liu
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; TCM Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Hao Liu
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Junhua Zhang
- TCM Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Han Zhang
- TCM Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Yingchao Wang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Mirko Baruscotti
- Department of Bioscienze, Pacelab, University of Milano, Milan, Italy
| | - Lu Zhao
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yi Wang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
21
|
Jiang H, Esparza TJ, Kummer TT, Zhong H, Rettig J, Brody DL. Live Neuron High-Content Screening Reveals Synaptotoxic Activity in Alzheimer Mouse Model Homogenates. Sci Rep 2020; 10:3412. [PMID: 32098978 PMCID: PMC7042280 DOI: 10.1038/s41598-020-60118-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 02/05/2020] [Indexed: 12/28/2022] Open
Abstract
Accurate quantification of synaptic changes is essential for understanding the molecular mechanisms of synaptogenesis, synaptic plasticity, and synaptic toxicity. Here we demonstrate a robust high-content imaging method for the assessment of synaptic changes and apply the method to brain homogenates from an Alzheimer's disease mouse model. Our method uses serial imaging of endogenous fluorescent labeled presynaptic VAMP2 and postsynaptic PSD95 in long-term cultured live primary neurons in 96 well microplates, and uses automatic image analysis to quantify the number of colocalized mature synaptic puncta for the assessment of synaptic changes in live neurons. As a control, we demonstrated that our synaptic puncta assay is at least 10-fold more sensitive to the toxic effects of glutamate than the MTT assay. Using our assay, we have compared synaptotoxic activities in size-exclusion chromatography fractioned protein samples from 3xTg-AD mouse model brain homogenates. Multiple synaptotoxic activities were found in high and low molecular weight fractions. Amyloid-beta immunodepletion alleviated some but not all of the synaptotoxic activities. Although the biochemical entities responsible for the synaptotoxic activities have yet to be determined, these proof-of-concept results demonstrate that this novel assay may have many potential mechanistic and therapeutic applications.
Collapse
Affiliation(s)
- Hao Jiang
- Department of Neurology, Washington University School of Medicine, 660 South Euclid Avenue, Box 8111, St Louis, Missouri, 63110, USA
| | - Thomas J Esparza
- Department of Neurology, Washington University School of Medicine, 660 South Euclid Avenue, Box 8111, St Louis, Missouri, 63110, USA
- Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, 20817, USA
- National Institute of Neurological Disorders and Stroke, 10 Center Drive, Bethesda, Maryland, 20892, USA
| | - Terrance T Kummer
- Department of Neurology, Washington University School of Medicine, 660 South Euclid Avenue, Box 8111, St Louis, Missouri, 63110, USA
| | - Haining Zhong
- Vollum Institute, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, Portland, Oregon, 97239, USA
| | - Jens Rettig
- Department of Physiology, Saarland University, Center for Integrative Physiology and Molecular Medicine (CIPMM), Building 48, Homburg, 66421, Germany
| | - David L Brody
- Department of Neurology, Washington University School of Medicine, 660 South Euclid Avenue, Box 8111, St Louis, Missouri, 63110, USA.
- National Institute of Neurological Disorders and Stroke, 10 Center Drive, Bethesda, Maryland, 20892, USA.
- Department of Neurology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, 20814, USA.
| |
Collapse
|
22
|
Anania MC, Di Marco T, Mazzoni M, Greco A. Targeting Non-Oncogene Addiction: Focus on Thyroid Cancer. Cancers (Basel) 2020; 12:cancers12010129. [PMID: 31947935 PMCID: PMC7017043 DOI: 10.3390/cancers12010129] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 12/21/2019] [Accepted: 12/24/2019] [Indexed: 12/12/2022] Open
Abstract
Thyroid carcinoma (TC) is the most common malignancy of endocrine organs with an increasing incidence in industrialized countries. The majority of TC are characterized by a good prognosis, even though cases with aggressive forms not cured by standard therapies are also present. Moreover, target therapies have led to low rates of partial response and prompted the emergence of resistance, indicating that new therapies are needed. In this review, we summarize current literature about the non-oncogene addiction (NOA) concept, which indicates that cancer cells, at variance with normal cells, rely on the activity of genes, usually not mutated or aberrantly expressed, essential for coping with the transformed phenotype. We highlight the potential of non-oncogenes as a point of intervention for cancer therapy in general, and present evidence for new putative non-oncogenes that are essential for TC survival and that may constitute attractive new therapeutic targets.
Collapse
|
23
|
Li S, Xia M. Review of high-content screening applications in toxicology. Arch Toxicol 2019; 93:3387-3396. [PMID: 31664499 PMCID: PMC7011178 DOI: 10.1007/s00204-019-02593-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 10/08/2019] [Indexed: 12/17/2022]
Abstract
High-content screening (HCS) technology combining automated microscopy and quantitative image analysis can address biological questions in academia and the pharmaceutical industry. Various HCS experimental applications have been utilized in the research field of in vitro toxicology. In this review, we describe several HCS application approaches used for studying the mechanism of compound toxicity, highlight some challenges faced in the toxicological community, and discuss the future directions of HCS in regards to new models, new reagents, data management, and informatics. Many specialized areas of toxicology including developmental toxicity, genotoxicity, developmental neurotoxicity/neurotoxicity, hepatotoxicity, cardiotoxicity, and nephrotoxicity will be examined. In addition, several newly developed cellular assay models including induced pluripotent stem cells (iPSCs), three-dimensional (3D) cell models, and tissues-on-a-chip will be discussed. New genome-editing technologies (e.g., CRISPR/Cas9), data analyzing tools for imaging, and coupling with high-content assays will be reviewed. Finally, the applications of machine learning to image processing will be explored. These new HCS approaches offer a huge step forward in dissecting biological processes, developing drugs, and making toxicology studies easier.
Collapse
Affiliation(s)
- Shuaizhang Li
- Division for Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD, USA
| | - Menghang Xia
- Division for Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD, USA.
| |
Collapse
|
24
|
Balaña-Fouce R, Pérez Pertejo MY, Domínguez-Asenjo B, Gutiérrez-Corbo C, Reguera RM. Walking a tightrope: drug discovery in visceral leishmaniasis. Drug Discov Today 2019; 24:1209-1216. [PMID: 30876846 DOI: 10.1016/j.drudis.2019.03.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 01/29/2019] [Accepted: 03/06/2019] [Indexed: 12/14/2022]
Abstract
The current commitment of the pharma industry, nongovernmental organizations and academia to find better treatments against neglected tropical diseases should end decades of challenge caused by these global scourges. The initial result of these efforts has been the introduction of enhanced combinations of drugs, currently in clinical use, or formulations thereof. Phenotypic screening based on intracellular parasite infections has been revealed as the first key tool of antileishmanial drug discovery, because most first-in-class drugs entering Phase I trials were discovered this way. The professional commitment among stakeholders has enabled the availability of a plethora of new chemical entities that fit the target product profile for these diseases. However, the rate of hit discovery in leishmaniasis is far behind that for other neglected diseases. This review defends the need to develop new screening methods that consider the part played not only by intracellular parasites but also by the host's immune system to generate disease-relevant assays and improve clinical outcomes.
Collapse
Affiliation(s)
- Rafael Balaña-Fouce
- Departamento de Ciencias Biomédicas, Universidad de León, Campus de Vegazana, E-24071 León, Spain
| | - M Yolanda Pérez Pertejo
- Departamento de Ciencias Biomédicas, Universidad de León, Campus de Vegazana, E-24071 León, Spain
| | - Bárbara Domínguez-Asenjo
- Departamento de Ciencias Biomédicas, Universidad de León, Campus de Vegazana, E-24071 León, Spain
| | - Camino Gutiérrez-Corbo
- Departamento de Ciencias Biomédicas, Universidad de León, Campus de Vegazana, E-24071 León, Spain
| | - Rosa M Reguera
- Departamento de Ciencias Biomédicas, Universidad de León, Campus de Vegazana, E-24071 León, Spain.
| |
Collapse
|
25
|
Puligedda RD, Sharma R, Al-Saleem FH, Kouiavskaia D, Velu AB, Kattala CD, Prendergast GC, Lynch DR, Chumakov K, Dessain SK. Capture and display of antibodies secreted by hybridoma cells enables fluorescent on-cell screening. MAbs 2019; 11:546-558. [PMID: 30794061 PMCID: PMC6512912 DOI: 10.1080/19420862.2019.1574520] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Hybridoma methods for monoclonal antibody (mAb) cloning are a mainstay of biomedical research, but they are hindered by the need to maintain hybridomas in oligoclonal pools during antibody screening. Here, we describe a system in which hybridomas specifically capture and display the mAbs they secrete: On-Cell mAb Screening (OCMS™). In OCMS™, mAbs displayed on the cell surface can be rapidly assayed for expression level and binding specificity using fluorescent antigens with high-content (image-based) methods or flow cytometry. OCMS™ demonstrated specific mAb binding to poliovirus and rabies virus by forming a cell surface IgG “cap”, as a universal assay for anti-viral mAbs. We produced and characterized OCMS™-enabled hybridomas secreting mAbs that neutralize poliovirus and used fluorescence microscopy to identify and clone a human mAb specific for the human N-methyl-D-aspartate receptor. Lastly, we used OCMS™ to assess expression and antigen binding of a recombinant mAb produced in 293T cells. As a novel method to physically associate mAbs with the hybridomas that secrete them, OCMS™ overcomes a central challenge to hybridoma mAb screening and offers new paradigms for mAb discovery and production.
Collapse
Affiliation(s)
| | - Rashmi Sharma
- a Lankenau Institute for Medical Research , Wynnewood , PA , USA
| | | | - Diana Kouiavskaia
- b Center for Biologics Evaluation and Research , Food and Drug Administration , Silver Spring , MD , USA
| | - Arul Balaji Velu
- a Lankenau Institute for Medical Research , Wynnewood , PA , USA
| | | | | | - David R Lynch
- c Division of Neurology , Children's Hospital of Pennsylvania , Philadelphia , PA , USA
| | - Konstantin Chumakov
- b Center for Biologics Evaluation and Research , Food and Drug Administration , Silver Spring , MD , USA
| | - Scott K Dessain
- a Lankenau Institute for Medical Research , Wynnewood , PA , USA
| |
Collapse
|
26
|
Apáti Á, Varga N, Berecz T, Erdei Z, Homolya L, Sarkadi B. Application of human pluripotent stem cells and pluripotent stem cell-derived cellular models for assessing drug toxicity. Expert Opin Drug Metab Toxicol 2018; 15:61-75. [PMID: 30526128 DOI: 10.1080/17425255.2019.1558207] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Introduction: Human pluripotent stem cells (hPSCs) are capable of differentiating into all types of cells in the body and so provide suitable toxicology screening systems even for hard-to-obtain human tissues. Since hPSCs can also be generated from differentiated cells and current gene editing technologies allow targeted genome modifications, hPSCs can be applied for drug toxicity screening both in normal and disease-specific models. Targeted hPSC differentiation is still a challenge but cardiac, neuronal or liver cells, and complex cellular models are already available for practical applications. Areas covered: The authors review new gene-editing and cell-biology technologies to generate sensitive toxicity screening systems based on hPSCs. Then the authors present the use of undifferentiated hPSCs for examining embryonic toxicity and discuss drug screening possibilities in hPSC-derived models. The authors focus on the application of human cardiomyocytes, hepatocytes, and neural cultures in toxicity testing, and discuss the recent possibilities for drug screening in a 'body-on-a-chip' model system. Expert opinion: hPSCs and their genetically engineered derivatives provide new possibilities to investigate drug toxicity in human tissues. The key issues in this regard are still the selection and generation of proper model systems, and the interpretation of the results in understanding in vivo drug effects.
Collapse
Affiliation(s)
- Ágota Apáti
- a Institute of Enzymology , Research Centre for Natural Sciences , Budapest , Hungary
| | - Nóra Varga
- a Institute of Enzymology , Research Centre for Natural Sciences , Budapest , Hungary
| | - Tünde Berecz
- a Institute of Enzymology , Research Centre for Natural Sciences , Budapest , Hungary
| | - Zsuzsa Erdei
- a Institute of Enzymology , Research Centre for Natural Sciences , Budapest , Hungary
| | - László Homolya
- a Institute of Enzymology , Research Centre for Natural Sciences , Budapest , Hungary
| | - Balázs Sarkadi
- a Institute of Enzymology , Research Centre for Natural Sciences , Budapest , Hungary
| |
Collapse
|
27
|
Yan X, Zhou L, Wu Z, Wang X, Chen X, Yang F, Guo Y, Wu M, Chen Y, Li W, Wang J, Du Y. High throughput scaffold-based 3D micro-tumor array for efficient drug screening and chemosensitivity testing. Biomaterials 2018; 198:167-179. [PMID: 29807624 DOI: 10.1016/j.biomaterials.2018.05.020] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 05/04/2018] [Accepted: 05/13/2018] [Indexed: 12/23/2022]
Abstract
Oncology drug development is greatly hampered by inefficient drug screening using 2D culture. Herein, we present ready-to-use micro-scaffolds in 384-well format to generate uniform 3D micro-tumor array (3D-MTA, CV < 0.15) that predicts in vivo drug responses more accurately than 2D monolayer. 3D-MTA generated from both cell lines and primary cells achieved high screen quality (Z' > 0.5), and were compatible with standard high throughput and high content instruments. Doxorubicin identified by 3D-MTA and 2D successfully inhibited tumor growth in mice bearing lung cancer cell line (H226) xenografts, but not gemcitabine and vinorelbine, which were selected solely by 2D. Resistance towards targeted therapy was modeled on 3D-MTA, which elicited SK-BR-3 to express higher proliferation-related genes in response to gefitinb, as compared to 2D. Screening of 56 MAPK inhibitors identified pisamertib to synergistically improve cytotoxicity effect in combination with gefitinib. Primary tumor cells derived from patient-derived xenografts further attested concordance of drug response in 3D-MTA with in vivo response. 3D-MTA was further extended to realize chemosensitivity testing using patient-derived cells. Overall, 3D-MTA demonstrated strong potential to accelerate drug discovery and improve cancer treatment by providing efficient drug screening.
Collapse
Affiliation(s)
- Xiaojun Yan
- Department of Biomedical Engineering, School of Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University, Beijing, 100084, PR China
| | - Lyu Zhou
- Department of Biomedical Engineering, School of Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University, Beijing, 100084, PR China; School of Life Sciences, Tsinghua University, Beijing, 100084, PR China
| | - Zhaozhao Wu
- Department of Biomedical Engineering, School of Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University, Beijing, 100084, PR China
| | - Xun Wang
- Department of Thoracic Surgery, Peking University People's Hospital, Peking University, Beijing, 100044, PR China
| | - Xiuyuan Chen
- Department of Thoracic Surgery, Peking University People's Hospital, Peking University, Beijing, 100044, PR China
| | - Fan Yang
- Department of Thoracic Surgery, Peking University People's Hospital, Peking University, Beijing, 100044, PR China
| | - Yanan Guo
- Beijing Biocytogen Co., Ltd, Beijing, 100176, PR China
| | - Min Wu
- Beijing Biocytogen Co., Ltd, Beijing, 100176, PR China
| | - Yuyang Chen
- Department of Biomedical Engineering, School of Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University, Beijing, 100084, PR China
| | - Wenjing Li
- Department of Biomedical Engineering, School of Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University, Beijing, 100084, PR China
| | - Jun Wang
- Department of Thoracic Surgery, Peking University People's Hospital, Peking University, Beijing, 100044, PR China.
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University, Beijing, 100084, PR China.
| |
Collapse
|
28
|
Wilson AC, Moutsatsos IK, Yu G, Pineda JJ, Feng Y, Auld DS. A Scalable Pipeline for High-Throughput Flow Cytometry. SLAS DISCOVERY 2018; 23:708-718. [PMID: 29768981 DOI: 10.1177/2472555218774770] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Flow cytometry (FC) provides high-content data for a variety of applications, including phenotypic analysis of cell surface and intracellular markers, characterization of cell supernatant or lysates, and gene expression analysis. Historically, sample preparation, acquisition, and analysis have presented as a bottleneck for running such types of assays at scale. This article will outline the solutions that have been implemented at Novartis which have allowed high-throughput FC to be successfully conducted and analyzed for a variety of cell-based assays. While these experiments were generally conducted to measure phenotypic responses from a well-characterized and information-rich small molecular probe library known as the Mechanism-of-Action (MoA) Box, they are broadly applicable to any type of test sample. The article focuses on application of automated methods for FC sample preparation in 384-well assay plates. It also highlights a pipeline for analyzing large volumes of FC data, covering a visualization approach that facilitates review of screen-level data by dynamically embedding FlowJo (FJ) workspace images for each sample into a Spotfire file, directly linking them to the metric being observed. Finally, an application of these methods to a screen for MHC-I expression upregulators is discussed.
Collapse
Affiliation(s)
- Aaron C Wilson
- 1 Novartis Institutes of Biomedical Research, Cambridge, MA, USA
| | | | - Gary Yu
- 1 Novartis Institutes of Biomedical Research, Cambridge, MA, USA
| | - Javier J Pineda
- 2 Department Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Yan Feng
- 1 Novartis Institutes of Biomedical Research, Cambridge, MA, USA
| | - Douglas S Auld
- 1 Novartis Institutes of Biomedical Research, Cambridge, MA, USA
| |
Collapse
|
29
|
Zhang J, Jin Q, Li S, Wu J, Wang Z, Hou J, Qu H, Long H, Wu W, Guo D. Orientalol L–P, novel sesquiterpenes from the rhizome of Alisma orientale (Sam.) Juzep and their nephrotoxicity on HK2 cells. NEW J CHEM 2018. [DOI: 10.1039/c8nj02027b] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Two new sesquiterpenes and three new nor-sesquiterpenes from the rhizome of Alisma orientale (Sam.) Juzep, and their nephrotoxicities.
Collapse
Affiliation(s)
- Jianqing Zhang
- School of Pharmacy
- Shenyang Pharmaceutical University
- Shenyang 110016
- China
- Shanghai Institute of Materia Medica
| | - Qinghao Jin
- Shanghai Institute of Materia Medica
- Chinese Academy of Sciences
- Shanghai 201203
- China
| | - Shiyou Li
- Key Laboratory of Genomic and Precision Medicine
- Beijing Institute of Genomics
- Chinese Academy of Sciences
- Beijing 100101
- China
| | - Jia Wu
- Shanghai Institute of Materia Medica
- Chinese Academy of Sciences
- Shanghai 201203
- China
| | - Zhen Wang
- Key Laboratory of Genomic and Precision Medicine
- Beijing Institute of Genomics
- Chinese Academy of Sciences
- Beijing 100101
- China
| | - Jinjun Hou
- Shanghai Institute of Materia Medica
- Chinese Academy of Sciences
- Shanghai 201203
- China
| | - Hua Qu
- Shanghai Institute of Materia Medica
- Chinese Academy of Sciences
- Shanghai 201203
- China
| | - Huali Long
- Shanghai Institute of Materia Medica
- Chinese Academy of Sciences
- Shanghai 201203
- China
| | - Wanying Wu
- Shanghai Institute of Materia Medica
- Chinese Academy of Sciences
- Shanghai 201203
- China
| | - Dean Guo
- School of Pharmacy
- Shenyang Pharmaceutical University
- Shenyang 110016
- China
- Shanghai Institute of Materia Medica
| |
Collapse
|
30
|
Berggren E, White A, Ouedraogo G, Paini A, Richarz AN, Bois FY, Exner T, Leite S, Grunsven LAV, Worth A, Mahony C. Ab initio chemical safety assessment: A workflow based on exposure considerations and non-animal methods. COMPUTATIONAL TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2017; 4:31-44. [PMID: 29214231 PMCID: PMC5695905 DOI: 10.1016/j.comtox.2017.10.001] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 10/09/2017] [Accepted: 10/10/2017] [Indexed: 12/12/2022]
Abstract
We describe and illustrate a workflow for chemical safety assessment that completely avoids animal testing. The workflow, which was developed within the SEURAT-1 initiative, is designed to be applicable to cosmetic ingredients as well as to other types of chemicals, e.g. active ingredients in plant protection products, biocides or pharmaceuticals. The aim of this work was to develop a workflow to assess chemical safety without relying on any animal testing, but instead constructing a hypothesis based on existing data, in silico modelling, biokinetic considerations and then by targeted non-animal testing. For illustrative purposes, we consider a hypothetical new ingredient x as a new component in a body lotion formulation. The workflow is divided into tiers in which points of departure are established through in vitro testing and in silico prediction, as the basis for estimating a safe external dose in a repeated use scenario. The workflow includes a series of possible exit (decision) points, with increasing levels of confidence, based on the sequential application of the Threshold of Toxicological (TTC) approach, read-across, followed by an "ab initio" assessment, in which chemical safety is determined entirely by new in vitro testing and in vitro to in vivo extrapolation by means of mathematical modelling. We believe that this workflow could be applied as a tool to inform targeted and toxicologically relevant in vitro testing, where necessary, and to gain confidence in safety decision making without the need for animal testing.
Collapse
Affiliation(s)
- Elisabet Berggren
- Chemical Safety and Alternative Methods Unit, & EURL ECVAM, Directorate F – Health, Consumers and Reference Materials, Joint Research Centre, European Commission, Ispra, Italy
| | | | | | - Alicia Paini
- Chemical Safety and Alternative Methods Unit, & EURL ECVAM, Directorate F – Health, Consumers and Reference Materials, Joint Research Centre, European Commission, Ispra, Italy
| | - Andrea-Nicole Richarz
- Chemical Safety and Alternative Methods Unit, & EURL ECVAM, Directorate F – Health, Consumers and Reference Materials, Joint Research Centre, European Commission, Ispra, Italy
| | | | | | - Sofia Leite
- Liver Cell Biology Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
| | - Leo A. van Grunsven
- Liver Cell Biology Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
| | - Andrew Worth
- Chemical Safety and Alternative Methods Unit, & EURL ECVAM, Directorate F – Health, Consumers and Reference Materials, Joint Research Centre, European Commission, Ispra, Italy
| | | |
Collapse
|
31
|
Gul S. Epigenetic assays for chemical biology and drug discovery. Clin Epigenetics 2017; 9:41. [PMID: 28439316 PMCID: PMC5399855 DOI: 10.1186/s13148-017-0342-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Accepted: 04/12/2017] [Indexed: 12/27/2022] Open
Abstract
The implication of epigenetic abnormalities in many diseases and the approval of a number of compounds that modulate specific epigenetic targets in a therapeutically relevant manner in cancer specifically confirms that some of these targets are druggable by small molecules. Furthermore, a number of compounds are currently in clinical trials for other diseases including cardiovascular, neurological and metabolic disorders. Despite these advances, the approved treatments for cancer only extend progression-free survival for a relatively short time and being associated with significant side effects. The current clinical trials involving the next generation of epigenetic drugs may address the disadvantages of the currently approved epigenetic drugs. The identification of chemical starting points of many drugs often makes use of screening in vitro assays against libraries of synthetic or natural products. These assays can be biochemical (using purified protein) or cell-based (using for example, genetically modified, cancer cell lines or primary cells) and performed in microtiter plates, thus enabling a large number of samples to be tested. A considerable number of such assays are available to monitor epigenetic target activity, and this review provides an overview of drug discovery and chemical biology and describes assays that monitor activities of histone deacetylase, lysine-specific demethylase, histone methyltransferase, histone acetyltransferase and bromodomain. It is of critical importance that an appropriate assay is developed and comprehensively validated for a given drug target prior to screening in order to improve the probability of the compound progressing in the drug discovery value chain.
Collapse
Affiliation(s)
- Sheraz Gul
- Fraunhofer Institute for Molecular Biology and Applied Ecology - ScreeningPort, Schnackenburgallee 114, 22525 Hamburg, Germany
| |
Collapse
|
32
|
Martucci H, Campit SE, Gee SR, Bray WM, Gokey T, Cada AK, Yen TY, Minoura K, Guliaev AB, Lokey RS, Amagata T. Naphthablins B and C, Meroterpenoids Identified from the Marine Sediment-Derived Streptomyces sp. CP26-58 Using HeLa Cell-Based Cytological Profiling. JOURNAL OF NATURAL PRODUCTS 2017; 80:684-691. [PMID: 28128950 DOI: 10.1021/acs.jnatprod.6b00996] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
HeLa cell-based cytological profiling (CP) was applied to an extract library of marine sediment-derived actinomycetes to discover new cytotoxic secondary metabolites. Among the hit strains, Streptomyces sp. CP26-58 was selected for further investigation to identify its cytotoxic metabolites. CP revealed that the known ionophore tetronasin (1) was responsible for the cytotoxic effect found in the extract. Furthermore, three naphthoquinone meroterpenoids, naphthablin A (2) and two new derivatives designated as naphthablins B (3) and C (4), were isolated from other cytotoxic fractions. The structures of the new compounds were elucidated based on analysis of their HRESIMS and comprehensive NMR data. The absolute configurations of the new compounds were deduced by simulating ECD spectra and calculating potential energies for the model compounds using density function theory (DFT) calculations. Compound 1 showed a significant cytotoxic effect against HeLa cells with an IC50 value of 0.23 μM, and CP successfully clustered 1 with calcium ionophores.
Collapse
Affiliation(s)
- Hana Martucci
- Department of Chemistry and Biochemistry, San Francisco State University , 1600 Holloway Avenue, San Francisco, California 94132, United States
| | - Scott E Campit
- Department of Chemistry and Biochemistry, San Francisco State University , 1600 Holloway Avenue, San Francisco, California 94132, United States
| | - Stephanie R Gee
- Department of Chemistry and Biochemistry, San Francisco State University , 1600 Holloway Avenue, San Francisco, California 94132, United States
| | - Walter M Bray
- Department of Chemistry and Biochemistry, University of California, Santa Cruz , 1156 High Street, Santa Cruz, California, 95064, United States
| | - Trevor Gokey
- Department of Chemistry and Biochemistry, San Francisco State University , 1600 Holloway Avenue, San Francisco, California 94132, United States
| | - A King Cada
- Department of Chemistry and Biochemistry, San Francisco State University , 1600 Holloway Avenue, San Francisco, California 94132, United States
| | - Ten-Yang Yen
- Department of Chemistry and Biochemistry, San Francisco State University , 1600 Holloway Avenue, San Francisco, California 94132, United States
| | - Katsuhiko Minoura
- Osaka University of Pharmaceutical Sciences , 4-20-1 Nasahara, Takatsuki, Osaka 569-1094, Japan
| | - Anton B Guliaev
- Department of Chemistry and Biochemistry, San Francisco State University , 1600 Holloway Avenue, San Francisco, California 94132, United States
| | - R Scott Lokey
- Department of Chemistry and Biochemistry, University of California, Santa Cruz , 1156 High Street, Santa Cruz, California, 95064, United States
| | - Taro Amagata
- Department of Chemistry and Biochemistry, San Francisco State University , 1600 Holloway Avenue, San Francisco, California 94132, United States
| |
Collapse
|
33
|
Hosoya M, Czysz K. Translational Prospects and Challenges in Human Induced Pluripotent Stem Cell Research in Drug Discovery. Cells 2016; 5:cells5040046. [PMID: 28009813 PMCID: PMC5187530 DOI: 10.3390/cells5040046] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 11/27/2016] [Accepted: 12/15/2016] [Indexed: 02/07/2023] Open
Abstract
Despite continuous efforts to improve the process of drug discovery and development, achieving success at the clinical stage remains challenging because of a persistent translational gap between the preclinical and clinical settings. Under these circumstances, the discovery of human induced pluripotent stem (iPS) cells has brought new hope to the drug discovery field because they enable scientists to humanize a variety of pharmacological and toxicological models in vitro. The availability of human iPS cell-derived cells, particularly as an alternative for difficult-to-access tissues and organs, is increasing steadily; however, their use in the field of translational medicine remains challenging. Biomarkers are an essential part of the translational effort to shift new discoveries from bench to bedside as they provide a measurable indicator with which to evaluate pharmacological and toxicological effects in both the preclinical and clinical settings. In general, during the preclinical stage of the drug development process, in vitro models that are established to recapitulate human diseases are validated by using a set of biomarkers; however, their translatability to a clinical setting remains problematic. This review provides an overview of current strategies for human iPS cell-based drug discovery from the perspective of translational research, and discusses the importance of early consideration of clinically relevant biomarkers.
Collapse
Affiliation(s)
- Masaki Hosoya
- Integrated Technology Research Laboratories, Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan.
| | - Katherine Czysz
- Integrated Technology Research Laboratories, Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan.
| |
Collapse
|
34
|
Donato MT, Gómez-Lechón MJ, Tolosa L. Using high-content screening technology for studying drug-induced hepatotoxicity in preclinical studies. Expert Opin Drug Discov 2016; 12:201-211. [DOI: 10.1080/17460441.2017.1271784] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Maria Teresa Donato
- Unidad de Hepatología Experimental, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
- Fondo de Investigaciones Sanitarias, CIBEREHD, Madrid, Spain
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | - Maria José Gómez-Lechón
- Unidad de Hepatología Experimental, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
- Fondo de Investigaciones Sanitarias, CIBEREHD, Madrid, Spain
| | - Laia Tolosa
- Unidad de Hepatología Experimental, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| |
Collapse
|
35
|
Stengl A, Hörl D, Leonhardt H, Helma J. A Simple and Sensitive High-Content Assay for the Characterization of Antiproliferative Therapeutic Antibodies. SLAS DISCOVERY 2016; 22:309-315. [PMID: 27909235 PMCID: PMC5322830 DOI: 10.1177/1087057116677821] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Monoclonal antibodies (mAbs) have become a central class of therapeutic agents in particular as antiproliferative compounds. Their often complex modes of action require sensitive assays during early, functional characterization. Current cell-based proliferation assays often detect metabolites that are indicative of metabolic activity but do not directly account for cell proliferation. Measuring DNA replication by incorporation of base analogues such as 5-bromo-2'-deoxyuridine (BrdU) fills this analytical gap but was previously restricted to bulk effect characterization in enzyme-linked immunosorbent assay formats. Here, we describe a cell-based assay format for the characterization of antiproliferative mAbs regarding potency and mode of action in a single experiment. The assay makes use of single cell-based high-content-analysis (HCA) for the reliable quantification of replicating cells and DNA content via 5-ethynyl-2'-deoxyuridine (EdU) and 4',6-diamidino-2-phenylindole (DAPI), respectively, as sensitive measures of antiproliferative mAb activity. We used trastuzumab, an antiproliferative therapeutic antibody interfering with HER2 cell surface receptor-mediated growth signal transduction, and HER2-overexpressing cell lines BT474 and SKBR3 to demonstrate up to 10-fold signal-to-background (S/B) ratios for treated versus untreated cells and a shift in cell cycle profiles indicating antibody-induced cell cycle arrest. The assay is simple, cost-effective, and sensitive, providing a cell-based format for preclinical characterization of therapeutic mAbs.
Collapse
Affiliation(s)
- Andreas Stengl
- 1 Department of Biology II, LMU Munich, Planegg-Martinsried, Germany
| | - David Hörl
- 1 Department of Biology II, LMU Munich, Planegg-Martinsried, Germany
| | | | - Jonas Helma
- 1 Department of Biology II, LMU Munich, Planegg-Martinsried, Germany
| |
Collapse
|