1
|
Sheedy A, Burduli N, Prakash A, Gurney M, Hanley S, Prendeville H, Sarkar S, O'Dwyer J, O'Dwyer M, Dolan E. NK cell line modified to express a potent, DR5 specific variant of TRAIL, show enhanced cytotoxicity in ovarian cancer models. Heliyon 2024; 10:e34976. [PMID: 39170449 PMCID: PMC11336271 DOI: 10.1016/j.heliyon.2024.e34976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 07/15/2024] [Accepted: 07/19/2024] [Indexed: 08/23/2024] Open
Abstract
Objective Ovarian cancer is a lethal gynaecological malignancy with unsatisfactory 5 year survival rates of 30-50 %. Cell immunotherapy is a promising new cancer treatment where immune cells, such as Natural Killer (NK) cells, are administered to enable the patient to fight cancer through direct cytotoxicity. NK cells orchestrate an adaptive immune response by enabling the release of tumour antigens. NK cell cytotoxicity and effector responses are largely driven by TRAIL engagement. In this study we investigated the cytotoxic potential of a human NK cell line that were modified to express a potent DR5 specific TRAIL variant. We hypothesised that this modification would enhance NK cell cytotoxicity against TRAIL sensitive and resistant ovarian cancer cell lines in vitro. Methods KHYG-1 human NK cells were modified with a TRAIL variant targeting DR5 (TRAILv-KHYG-1). Human ovarian cancer cell lines, OVCAR-3 and SKOV-3, were cultured with modified or non-modified NK cells at different effector:target (E:T) ratios for 4 or 16 h. Apoptosis was assessed by Annexin-APC and 7-AAD and measured using flow cytometry. Apoptotic cells were defined as annexin V 7-AAD double positive. Cytokine expression was measured by multiplex ELISA, and analysed by flow cytometry. Results Modified and non-modified NK cells significantly reduced OVCAR-3 cell viability as compared to OVCAR-3 cells that were cultured alone after 4 and 16 h treatment. OVCAR-3 cell viability was reduced after treatment with 1:1 E:T ratio with TRAILv-KHYG-1 cells after 16 h. On the contrary, neither NK cell line had any effect of SKOV-3 cell viability despite SKOV-3 cells having more DR5 surface expression compared to OVCAR-3 cells. Conclusions TRAILv-KHYG-1 cells significantly reduced OVCAR-3 cell viability as compared to non-modified NK cells. However, no significant reduction in viability was observed when SKOV-3 cell were cultured with either NK cells, despite having more DR5 surface expression compared to OVCAR-3 cells. These data indicate that mechanisms other than DR5 expression drive TRAIL resistance in ovarian cancer.
Collapse
Affiliation(s)
- A.M. Sheedy
- Biomedical Engineering, School of Engineering, College of Science and Engineering, University of Galway, Ireland
- CÚRAM, Centre for Research in Medical Devices, University of Galway, Galway, Ireland
| | - N. Burduli
- Apoptosis Research Centre, University of Galway, Galway, Ireland
- Center for Hematology Regenerative Medicine (HERM), Karolinska Institutet, Stockholm, Sweden
| | - A. Prakash
- Biomedical Engineering, School of Engineering, College of Science and Engineering, University of Galway, Ireland
| | - M. Gurney
- Apoptosis Research Centre, University of Galway, Galway, Ireland
| | - S. Hanley
- Flow Cytometry Core Facility, University of Galway, Galway, Ireland
| | - H. Prendeville
- Biomedical Engineering, School of Engineering, College of Science and Engineering, University of Galway, Ireland
| | - S. Sarkar
- ONK Therapeutics Inc, Galway, Ireland
| | - J. O'Dwyer
- Biomedical Engineering, School of Engineering, College of Science and Engineering, University of Galway, Ireland
| | - M. O'Dwyer
- Apoptosis Research Centre, University of Galway, Galway, Ireland
- ONK Therapeutics Inc, Galway, Ireland
| | - E.B. Dolan
- Biomedical Engineering, School of Engineering, College of Science and Engineering, University of Galway, Ireland
- CÚRAM, Centre for Research in Medical Devices, University of Galway, Galway, Ireland
| |
Collapse
|
2
|
Cao M, Carlson RD, Staudt RE, Snook AE. In vitro assays to evaluate CAR-T cell cytotoxicity. Methods Cell Biol 2023; 183:303-315. [PMID: 38548415 DOI: 10.1016/bs.mcb.2023.05.009] [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] [Indexed: 04/02/2024]
Abstract
This chapter introduces four commonly used in vitro chimeric antigen receptor (CAR)-T cell cytotoxicity assays (lactate dehydrogenase release assay, 51Cr release assay, IncuCyte live cell killing assay, and xCELLigence real-time analysis) and provides a detailed protocol for xCELLigence real-time analysis. Focusing on in vitro assays, this chapter starts with explaining the mechanisms and discussing the utilization of each assay to quantify T-cell-induced cytotoxicity. Due to the high-throughput quantification and straightforward workflow of xCELLigence real-time analysis, a protocol entailing reagents and equipment, a 3-day step-by-step procedure, and instructions for data analysis are provided.
Collapse
Affiliation(s)
- Miao Cao
- Department of Pharmacology, Physiology, & Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Robert D Carlson
- Department of Pharmacology, Physiology, & Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Ross E Staudt
- Department of Pharmacology, Physiology, & Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Adam E Snook
- Department of Pharmacology, Physiology, & Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States; Department of Microbiology & Immunology, Thomas Jefferson University, Philadelphia, PA, United States; Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States.
| |
Collapse
|
3
|
Mensali N, Köksal H, Joaquina S, Wernhoff P, Casey NP, Romecin P, Panisello C, Rodriguez R, Vimeux L, Juzeniene A, Myhre MR, Fåne A, Ramírez CC, Maggadottir SM, Duru AD, Georgoudaki AM, Grad I, Maturana AD, Gaudernack G, Kvalheim G, Carcaboso AM, de Alava E, Donnadieu E, Bruland ØS, Menendez P, Inderberg EM, Wälchli S. ALPL-1 is a target for chimeric antigen receptor therapy in osteosarcoma. Nat Commun 2023; 14:3375. [PMID: 37291203 PMCID: PMC10250459 DOI: 10.1038/s41467-023-39097-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 05/25/2023] [Indexed: 06/10/2023] Open
Abstract
Osteosarcoma (OS) remains a dismal malignancy in children and young adults, with poor outcome for metastatic and recurrent disease. Immunotherapies in OS are not as promising as in some other cancer types due to intra-tumor heterogeneity and considerable off-target expression of the potentially targetable proteins. Here we show that chimeric antigen receptor (CAR) T cells could successfully target an isoform of alkaline phosphatase, ALPL-1, which is highly and specifically expressed in primary and metastatic OS. The target recognition element of the second-generation CAR construct is based on two antibodies, previously shown to react against OS. T cells transduced with these CAR constructs mediate efficient and effective cytotoxicity against ALPL-positive cells in in vitro settings and in state-of-the-art in vivo orthotopic models of primary and metastatic OS, without unexpected toxicities against hematopoietic stem cells or healthy tissues. In summary, CAR-T cells targeting ALPL-1 show efficiency and specificity in treating OS in preclinical models, paving the path for clinical translation.
Collapse
Affiliation(s)
- Nadia Mensali
- Translational Research Unit, Department of Cellular Therapy, Oslo University Hospital, Oslo, Norway
| | - Hakan Köksal
- Translational Research Unit, Department of Cellular Therapy, Oslo University Hospital, Oslo, Norway
| | - Sandy Joaquina
- Translational Research Unit, Department of Cellular Therapy, Oslo University Hospital, Oslo, Norway
| | - Patrik Wernhoff
- Translational Research Unit, Department of Cellular Therapy, Oslo University Hospital, Oslo, Norway
| | - Nicholas P Casey
- Translational Research Unit, Department of Cellular Therapy, Oslo University Hospital, Oslo, Norway
| | - Paola Romecin
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
- Red Española de Terapias Avanzadas (TERAV)-Instituto de Salud Carlos III (ISCIII) (RICORS, RD21/0017/0029), Madrid, Spain
| | - Carla Panisello
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
- Red Española de Terapias Avanzadas (TERAV)-Instituto de Salud Carlos III (ISCIII) (RICORS, RD21/0017/0029), Madrid, Spain
| | - René Rodriguez
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
- Centro de Investigación Biomédica en Red-Oncología (CIBER-ONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Lene Vimeux
- Université de Paris, Institut Cochin, INSERM, CNRS, Equipe labellisée Ligue Contre le Cancer, F-75014, PARIS, France
| | - Asta Juzeniene
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Marit R Myhre
- Translational Research Unit, Department of Cellular Therapy, Oslo University Hospital, Oslo, Norway
| | - Anne Fåne
- Translational Research Unit, Department of Cellular Therapy, Oslo University Hospital, Oslo, Norway
| | - Carolina Castilla Ramírez
- Institute of Biomedicine of Sevilla (IBiS), Virgen del Rocio University Hospital, CSIC, University of Sevilla, CIBER-ONC, 41013, Seville, Spain
| | | | - Adil Doganay Duru
- NSU Cell Therapy Institute, Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Anna-Maria Georgoudaki
- NSU Cell Therapy Institute, Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, USA
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Iwona Grad
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Andrés Daniel Maturana
- Laboratory of Animal Cell Physiology, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Gustav Gaudernack
- Department of Cancer Immunology, Oslo University Hospital, Oslo, Norway
| | - Gunnar Kvalheim
- Translational Research Unit, Department of Cellular Therapy, Oslo University Hospital, Oslo, Norway
| | - Angel M Carcaboso
- SJD Pediatric Cancer Center Barcelona, Institut de Recerca Sant Joan de Deu, Barcelona, 08950, Spain
| | - Enrique de Alava
- Institute of Biomedicine of Sevilla (IBiS), Virgen del Rocio University Hospital, CSIC, University of Sevilla, CIBER-ONC, 41013, Seville, Spain
- Department of Normal and Pathological Cytology and Histology, School of Medicine, University of Seville, 41009, Seville, Spain
| | - Emmanuel Donnadieu
- Université de Paris, Institut Cochin, INSERM, CNRS, Equipe labellisée Ligue Contre le Cancer, F-75014, PARIS, France
| | - Øyvind S Bruland
- Department of Oncology, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Pablo Menendez
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
- Red Española de Terapias Avanzadas (TERAV)-Instituto de Salud Carlos III (ISCIII) (RICORS, RD21/0017/0029), Madrid, Spain
- CIBER-ONC, ISCIII, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain
| | - Else Marit Inderberg
- Translational Research Unit, Department of Cellular Therapy, Oslo University Hospital, Oslo, Norway.
| | - Sébastien Wälchli
- Translational Research Unit, Department of Cellular Therapy, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
4
|
Zang X, Chen S, Zhu J, Ma J, Zhai Y. The Emerging Role of Central and Peripheral Immune Systems in Neurodegenerative Diseases. Front Aging Neurosci 2022; 14:872134. [PMID: 35547626 PMCID: PMC9082639 DOI: 10.3389/fnagi.2022.872134] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 03/25/2022] [Indexed: 12/31/2022] Open
Abstract
For decades, it has been widely believed that the blood-brain barrier (BBB) provides an immune privileged environment in the central nervous system (CNS) by blocking peripheral immune cells and humoral immune factors. This view has been revised in recent years, with increasing evidence revealing that the peripheral immune system plays a critical role in regulating CNS homeostasis and disease. Neurodegenerative diseases are characterized by progressive dysfunction and the loss of neurons in the CNS. An increasing number of studies have focused on the role of the connection between the peripheral immune system and the CNS in neurodegenerative diseases. On the one hand, peripherally released cytokines can cross the BBB, cause direct neurotoxicity and contribute to the activation of microglia and astrocytes. On the other hand, peripheral immune cells can also infiltrate the brain and participate in the progression of neuroinflammatory and neurodegenerative diseases. Neurodegenerative diseases have a high morbidity and disability rate, yet there are no effective therapies to stop or reverse their progression. In recent years, neuroinflammation has received much attention as a therapeutic target for many neurodegenerative diseases. In this review, we highlight the emerging role of the peripheral and central immune systems in neurodegenerative diseases, as well as their interactions. A better understanding of the emerging role of the immune systems may improve therapeutic strategies for neurodegenerative diseases.
Collapse
Affiliation(s)
- Xin Zang
- Department of Infectious Disease, Shengjing Hospital of China Medical University, Shenyang, China
| | - Si Chen
- Department of Neurology, the Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - JunYao Zhu
- Department of Infectious Disease, Shengjing Hospital of China Medical University, Shenyang, China
| | - Junwen Ma
- Department of Infectious Disease, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yongzhen Zhai
- Department of Infectious Disease, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
5
|
Salvo P, Vivaldi FM, Bonini A, Biagini D, Bellagambi FG, Miliani FM, Di Francesco F, Lomonaco T. Biosensors for Detecting Lymphocytes and Immunoglobulins. BIOSENSORS 2020; 10:E155. [PMID: 33121071 PMCID: PMC7694141 DOI: 10.3390/bios10110155] [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] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/23/2020] [Accepted: 10/26/2020] [Indexed: 12/15/2022]
Abstract
Lymphocytes (B, T and natural killer cells) and immunoglobulins are essential for the adaptive immune response against external pathogens. Flow cytometry and enzyme-linked immunosorbent (ELISA) kits are the gold standards to detect immunoglobulins, B cells and T cells, whereas the impedance measurement is the most used technique for natural killer cells. For point-of-care, fast and low-cost devices, biosensors could be suitable for the reliable, stable and reproducible detection of immunoglobulins and lymphocytes. In the literature, such biosensors are commonly fabricated using antibodies, aptamers, proteins and nanomaterials, whereas electrochemical, optical and piezoelectric techniques are used for detection. This review describes how these measurement techniques and transducers can be used to fabricate biosensors for detecting lymphocytes and the total content of immunoglobulins. The various methods and configurations are reported, along with the advantages and current limitations.
Collapse
Affiliation(s)
- Pietro Salvo
- Institute of Clinical Physiology, National Council of Research, Via Moruzzi 1, 56124 Pisa, Italy;
| | - Federico M. Vivaldi
- Institute of Clinical Physiology, National Council of Research, Via Moruzzi 1, 56124 Pisa, Italy;
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Moruzzi 13, 56124 Pisa, Italy; (A.B.); (D.B.); (F.M.M.); (F.D.F.); (T.L.)
| | - Andrea Bonini
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Moruzzi 13, 56124 Pisa, Italy; (A.B.); (D.B.); (F.M.M.); (F.D.F.); (T.L.)
| | - Denise Biagini
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Moruzzi 13, 56124 Pisa, Italy; (A.B.); (D.B.); (F.M.M.); (F.D.F.); (T.L.)
| | - Francesca G. Bellagambi
- Institut des Sciences Analytiques, UMR 5280, Université Lyon 1, 5, rue de la Doua, 69100 Villeurbanne, France;
| | - Filippo M. Miliani
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Moruzzi 13, 56124 Pisa, Italy; (A.B.); (D.B.); (F.M.M.); (F.D.F.); (T.L.)
| | - Fabio Di Francesco
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Moruzzi 13, 56124 Pisa, Italy; (A.B.); (D.B.); (F.M.M.); (F.D.F.); (T.L.)
| | - Tommaso Lomonaco
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Moruzzi 13, 56124 Pisa, Italy; (A.B.); (D.B.); (F.M.M.); (F.D.F.); (T.L.)
| |
Collapse
|
6
|
Yah CS, Simate GS. Engineered nanoparticle bio-conjugates toxicity screening: The xCELLigence cells viability impact. ACTA ACUST UNITED AC 2020; 10:195-203. [PMID: 32793442 PMCID: PMC7416007 DOI: 10.34172/bi.2020.24] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 12/09/2019] [Accepted: 12/21/2019] [Indexed: 12/23/2022]
Abstract
Introduction: The vast diverse products and applications of engineered nanoparticle bio-conjugates (ENPBCs) are increasing, and thus flooding the-markets. However, the data to support risk estimates of ENPBC are limited. While it is important to assess the potential benefits, acceptability and uptake, it is equally important to understand where ENPBCs safety is and how to expand and affirm consumer security concerns. Methods: Online articles were extracted from 2013 to 2016 that pragmatically used xCELLigence real-time cell analysis (RTCA) technology to describe the in-vitro toxicity of ENPBCs. The xCELLigence is a +noninvasive in vitro toxicity monitoring process that mimics exact continuous cellular bio-responses in real-time settings. On the other hand, articles were also extracted from 2008 to 2016 describing the in vivo animal models toxicity of ENPBCs with regards to safety outcomes. Results: Out of 32 of the 121 (26.4%) articles identified from the literature, 23 (71.9%) met the in-vitro xCELLigence and 9(28.1%) complied with the in vivo animal model toxicity inclusion criteria. Of the 23 articles, 4 of them (17.4%) had no size estimation of ENPBCs. The xCELLigence technology provided information on cell interactions, viability, and proliferation process. Eighty-three (19/23) of the in vitro xCELLigence technology studies described ENPBCs as nontoxic or partially nontoxic materials. The in vivo animal model provided further toxicity information where 1(1/9) of the in vivo animal model studies indicated potential animal toxicity while the remaining results recommended ENPPCs as potential candidates for drug therapy though with limited information on toxicity. Conclusion: The results showed that the bioimpacts of ENPBCs either at the in vitro or at in vivo animal model levels are still limited due to insufficient information and data. To keep pace with ENPBCs biomedical products and applications, in vitro, in vivo assays, clinical trials and long-term impacts are needed to validate their usability and uptake. Besides, more real-time ENPBCs-cell impact analyses using xCELLigence are needed to provide significant data and information for further in vivo testing.
Collapse
Affiliation(s)
- Clarence S Yah
- Implementation Science Unit, Wits Reproductive Health and HIV Institute, Faculty of Health Sciences, University of the Witwatersrand, South Africa.,School of Health Systems and Public Health, Faculty of Health Sciences, University of Pretoria, South Africa
| | - Geoffrey S Simate
- School of Chemical and Metallurgical Engineering, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
7
|
Gao W, Xu S, Zhang M, Liu S, Siu SPK, Peng H, Ng JCW, Tsao GSW, Chan AWH, Chow VLY, Chan JYW, Wong TS. NADPH oxidase 5α promotes the formation of CD271 tumor-initiating cells in oral cancer. Am J Cancer Res 2020; 10:1710-1727. [PMID: 32642285 PMCID: PMC7339284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 03/31/2020] [Indexed: 02/05/2023] Open
Abstract
Oral tongue squamous cell carcinoma (OTSCC) has a distinctive cell sub-population known as tumor-initiating cells (TICs). CD271 is a functional TIC receptor in head and neck cancers. The molecular mechanisms governing CD271 up-regulation remains unclear. Oxidative stress is a contributing factor in TIC development. Here, we explored the potential role of NADPH oxidase 5 (NOX5) and its regulatory mechanism on the development of CD271-expressing OTSCC. Our results showed that the splice variant NOX5α is the most prevalent form expressed in head and neck cancers. NOX5α enhanced OTSCC proliferation, migration, and invasion. Overexpression of NOX5α increased the size of OTSCC xenograft significantly in vivo. The tumor-promoting functions of NOX5α were mediated through the reactive oxygen species (ROS)-generating property. NOX5α activated ERK singling and increased CD271 expression at the transcription level. Also, NOX5α reduces the sensitivity of OTSCC to cisplatin and natural killer cells. The findings indicate that NOX5α plays an important part in the development of TIC in OTSCC.
Collapse
Affiliation(s)
- Wei Gao
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong21 Sassoon Road, Pokfulam, Hong Kong, China
| | - Shaowei Xu
- Department of Head and Neck Surgery, Cancer Hospital of Shantou University Medical College7 Raoping Road, Shantou 515031, Guangdong Province, China
| | - Minjuan Zhang
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong21 Sassoon Road, Pokfulam, Hong Kong, China
| | - Shuai Liu
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong21 Sassoon Road, Pokfulam, Hong Kong, China
| | - Sharie Pui-Kei Siu
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong21 Sassoon Road, Pokfulam, Hong Kong, China
| | - Hanwei Peng
- Department of Head and Neck Surgery, Cancer Hospital of Shantou University Medical College7 Raoping Road, Shantou 515031, Guangdong Province, China
| | - Judy Chun-Wai Ng
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong21 Sassoon Road, Pokfulam, Hong Kong, China
| | - George Sai-Wah Tsao
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong21 Sassoon Road, Pokfulam, Hong Kong, China
| | - Anthony Wing-Hung Chan
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong30-32 Ngan Shing Street, Shatin, NT, China
| | - Velda Ling-Yu Chow
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong21 Sassoon Road, Pokfulam, Hong Kong, China
| | - Jimmy Yu-Wai Chan
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong21 Sassoon Road, Pokfulam, Hong Kong, China
| | - Thian-Sze Wong
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong21 Sassoon Road, Pokfulam, Hong Kong, China
| |
Collapse
|
8
|
Lv D, Xu Y, Cheng H, Ke Y, Zhang X, Ying K. A novel cell-based assay for dynamically detecting neutrophil extracellular traps-induced lung epithelial injuries. Exp Cell Res 2020; 394:112101. [PMID: 32474064 PMCID: PMC7256615 DOI: 10.1016/j.yexcr.2020.112101] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 05/16/2020] [Accepted: 05/19/2020] [Indexed: 02/07/2023]
Abstract
Acute lung injury (ALI) and its more severe form, acute respiratory distress syndrome (ARDS) are common lung disorders characterized by alveolar-capillary barrier disruption and dyspnea, which can cause substantial morbidity and mortality. Currently, a cluster of acute respiratory illnesses, known as novel coronavirus (2019-nCoV)-infected pneumonia (NCIP), which allegedly originally occurred in Wuhan, China, has increased rapidly worldwide. The critically ill patients with ARDS have high mortality in subjects with comorbidities. Previously, the excessive recruitment and activation of neutrophils (polymorphonuclear leukocytes [PMNs]), accompanied by neutrophil extracellular traps (NETs) formation were reported being implicated in the pathogenesis of ALI/ARDS. However, the direct visualization of lung epithelial injuries caused by NETs, and the qualitative and quantitative evaluations of this damage are still lacking. Additionally, those already reported methods are limited for their neglect of the pathological role exerted by NETs and focusing only on the morphological features of NETosis. Therefore, we established a cell-based assay for detecting NETs during lung epithelial cells-neutrophils co-culture using the xCELLigence system, a recognized real-time, dynamic, label-free, sensitive, and high-throughput apparatus. Our results demonstrated that lung epithelial injuries, reflected by declines in cell index (CI) values, could be induced by lipopolysaccharide (LPS)-activated PMNs, or NETs in a time and dose-dependent manner. NETs generation was verified to be the major contributor to the cytotoxicity of activated PMNs; protein components of NETs were the prevailing cytotoxic mediators. Moreover, this cell-based assay identified that PMNs from severe pneumonia patients had a high NETs formative potential. Additionally, acetylsalicylic acid (ASA) and acetaminophen (APAP) were discovered alleviating NETs formation. Thus, this study not only presents a new methodology for detecting the pathophysiologic role of NETs but also lays down a foundation for exploring therapeutic interventions in an effort to cure ALI/ARDS in the clinical setting of severe pneumonia, including the emerging of NCIP. A real-time, dynamical and label-free assay for detecting NETs is established using the xCELLigence system. This establishment relies on the co-culture of lung epithelia and neutrophils, focusing on evaluating NETs’ effects. This cell-based assay has feasibility and practicality in clinical applications. This methodology builds a solid foundation for exploring therapies for ALI/ARDS, including the emerging NCIP.
Collapse
Affiliation(s)
- Dandan Lv
- Department of Respiratory Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, China
| | - Yiming Xu
- Department of Respiratory Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, China
| | - Hongqiang Cheng
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Yuehai Ke
- Department of Pathology and Pathophysiology and Department of Respiratory Medicine at Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Xue Zhang
- Department of Pathology and Pathophysiology and Department of Respiratory Medicine at Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China.
| | - Kejing Ying
- Department of Respiratory Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, China.
| |
Collapse
|
9
|
In-vitro analysis of free radical scavenging activities and suppression of LPS-induced ROS production in macrophage cells by Solanum sisymbriifolium extracts. Sci Rep 2020; 10:6493. [PMID: 32300192 PMCID: PMC7162848 DOI: 10.1038/s41598-020-63491-w] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 03/30/2020] [Indexed: 02/03/2023] Open
Abstract
The current study aims to evaluate the antioxidant, cytotoxicity activities and suppression of LPS-induced oxidative stress production and characterization of phytochemicals in Solanum sisymbriifolium leaf extracts. The 2,2-diphenyl-1-picrylhydrazyl (DPPH) and 2,2'-azino-bis3-ethylbenzothiazoline-6-sulphonic acid (ABTS) radical scavenging activity of the leaves of S. sisymbriifolium extracted with solvents of various polarities viz. water: ethanol, ratio 50: 50; ethyl acetate and dichloromethane, was assessed. The cytotoxicity of the extracts was determined using the [3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide] (MTT) assay on RAW 264.7 macrophage (Murine) cells and real-time cell analysis (RTCA) xCELLigence system was used for determining cell viability. Cell-based detection of reactive oxygen species (ROS) was investigated utilizing a 2',7'-Dichlorodihydrofluorescein diacetate (H2DCF-DA) assay. The DPPH and ABTS scavenging activity results of extracts revealed a dose-dependent response with significantly lower activity in both DPPH and ABTS. The superoxide dismutase (SOD) enzyme activity was then evaluated and extracts displayed a high SOD enzyme activity with 90-50% activity. Cytotoxicity results revealed that S. sisymbriifolium extracts were not toxic to RAW 264.7 macrophage cells at the tested concentrations. All three extracts decreased the production of ROS in macrophage cells. Phytochemical analysis using Fourier-transform infrared spectroscopy (FTIR) indicated the presence of metabolite functional groups which may be responsible for the antioxidant activity. The current study indicates that S. sisymbriifolium contains phytochemicals that scavenge free radicals, with less toxicity, and suppresses the LPS-induced ROS production in RAW 264.7 macrophage cells.
Collapse
|
10
|
Fasbender F, Obholzer M, Metzler S, Stöber R, Hengstler JG, Watzl C. Enhanced activation of human NK cells by drug-exposed hepatocytes. Arch Toxicol 2020; 94:439-448. [PMID: 32060585 DOI: 10.1007/s00204-020-02668-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 02/06/2020] [Indexed: 01/06/2023]
Abstract
Drug-induced liver injury (DILI) represents one of the major causes why drugs have to be withdrawn from the market. In this study, we describe a new interaction between drug-exposed hepatocytes and natural killer (NK) cells. In a previous genome-wide expression analysis of primary human hepatocytes that had been exposed to clinically relevant concentrations of 148 drugs, we found that several activating ligands for NK cell receptors were regulated by various drugs (e.g., valproic acid, ketoconazole, promethazine, isoniazid). Especially expression of the activating NKG2D ligands (MICA, MICB and ULBPs) and the NKp30 ligand B7-H6 were upregulated in primary human hepatocytes upon exposure to many different drugs. Using the human hepatocyte cell lines Huh7 and HepG2, we confirmed that protein levels of activating NK cell ligands were elevated after drug exposure. Hepatocyte cell lines or primary human hepatocytes co-cultivated with NK cells caused enhanced NK cell activation after pretreatment with drugs at in vivo relevant concentrations compared to solvent controls. Enhanced NK cell activation was evident by increased cytotoxicity against hepatocytes and interferon (IFN)-γ production. NK cell activation could be blocked by specific antibodies against activating NK cell receptors. These data support the hypothesis that NK cells can modulate drug-induced liver injury by direct interaction with hepatocytes resulting in cytotoxicity and IFN-γ production.
Collapse
Affiliation(s)
- Frank Fasbender
- Department of Immunology, Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany
| | - Martin Obholzer
- Department of Immunology, Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany
| | - Sarah Metzler
- Department of Immunology, Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany
- Department of Toxicology, Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany
| | - Regina Stöber
- Department of Toxicology, Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany
| | - Jan G Hengstler
- Department of Toxicology, Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany
| | - Carsten Watzl
- Department of Immunology, Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany.
| |
Collapse
|
11
|
Morgan K, Gamal W, Samuel K, Morley SD, Hayes PC, Bagnaninchi P, Plevris JN. Application of Impedance-Based Techniques in Hepatology Research. J Clin Med 2019; 9:jcm9010050. [PMID: 31878354 PMCID: PMC7019217 DOI: 10.3390/jcm9010050] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 12/18/2019] [Accepted: 12/19/2019] [Indexed: 12/22/2022] Open
Abstract
There are a variety of end-point assays and techniques available to monitor hepatic cell cultures and study toxicity within in vitro models. These commonly focus on one aspect of cell metabolism and are often destructive to cells. Impedance-based cellular assays (IBCAs) assess biological functions of cell populations in real-time by measuring electrical impedance, which is the resistance to alternating current caused by the dielectric properties of proliferating of cells. While the uses of IBCA have been widely reported for a number of tissues, specific uses in the study of hepatic cell cultures have not been reported to date. IBCA monitors cellular behaviour throughout experimentation non-invasively without labelling or damage to cell cultures. The data extrapolated from IBCA can be correlated to biological events happening within the cell and therefore may inform drug toxicity studies or other applications within hepatic research. Because tight junctions comprise the blood/biliary barrier in hepatocytes, there are major consequences when these junctions are disrupted, as many pathologies centre around the bile canaliculi and flow of bile out of the liver. The application of IBCA in hepatology provides a unique opportunity to assess cellular polarity and patency of tight junctions, vital to maintaining normal hepatic function. Here, we describe how IBCAs have been applied to measuring the effect of viral infection, drug toxicity /IC50, cholangiopathies, cancer metastasis and monitoring of the gut-liver axis. We also highlight key areas of research where IBCAs could be used in future applications within the field of hepatology.
Collapse
Affiliation(s)
- Katie Morgan
- The University of Edinburgh Hepatology Laboratory, Division of Heath Sciences, University of Edinburgh Medical School, Chancellor’s Building, Edinburgh BioQuarter, 49 Little France Crescent, Edinburgh EH16 4SB, UK; (S.D.M.); (P.C.H.); (J.N.P.)
- Correspondence:
| | - Wesam Gamal
- James Nasmyth Building, Institute of Mechanical, Process and Energy Engineering, Heriot-Watt University School of Engineering and Physical Sciences, Edinburgh EH14 4AS, UK;
| | - Kay Samuel
- The Jack Copland Centre, Advanced Therapeutics, Scottish National Blood Transfusion Service, 52 Research Avenue North, Edinburgh EH14 4BE, UK;
| | - Steven D. Morley
- The University of Edinburgh Hepatology Laboratory, Division of Heath Sciences, University of Edinburgh Medical School, Chancellor’s Building, Edinburgh BioQuarter, 49 Little France Crescent, Edinburgh EH16 4SB, UK; (S.D.M.); (P.C.H.); (J.N.P.)
| | - Peter C. Hayes
- The University of Edinburgh Hepatology Laboratory, Division of Heath Sciences, University of Edinburgh Medical School, Chancellor’s Building, Edinburgh BioQuarter, 49 Little France Crescent, Edinburgh EH16 4SB, UK; (S.D.M.); (P.C.H.); (J.N.P.)
| | - Pierre Bagnaninchi
- MRC Centre for Regenerative Medicine 5 Little France Drive, Edinburgh EH16 4UU, UK;
| | - John N. Plevris
- The University of Edinburgh Hepatology Laboratory, Division of Heath Sciences, University of Edinburgh Medical School, Chancellor’s Building, Edinburgh BioQuarter, 49 Little France Crescent, Edinburgh EH16 4SB, UK; (S.D.M.); (P.C.H.); (J.N.P.)
| |
Collapse
|
12
|
Johnson RH, Kho DT, O' Carroll SJ, Angel CE, Graham ES. The functional and inflammatory response of brain endothelial cells to Toll-Like Receptor agonists. Sci Rep 2018; 8:10102. [PMID: 29973684 PMCID: PMC6031625 DOI: 10.1038/s41598-018-28518-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 06/14/2018] [Indexed: 02/06/2023] Open
Abstract
Toll-Like receptors (TLRs) represent an important early warning mechanism for the immune system to detect infection or tissue damage. The focus of this research was to determine the neuroinflammatory responses to commercial TLR ligands and their effects on brain endothelial barrier strength. Using biosensor technology we screened TLR ligands to all human TLRs and found that the brain endothelial hCMVECs cell line only responded to Poly(I:C) (TLR3-ligand), LPS (TLR4-ligand) and Imiquimod (TLR7 ligand). Both Poly(I:C) and LPS induced pronounced pro-inflammatory cytokine secretion as expected, whereas Imiquimod did not induce secretion of any pro-inflammatory cytokines. Using ECIS technology to measure endothelial barrier function, LPS and Poly(I:C) both acutely reduced barrier-strength, whereas Imiquimod caused immediate and sustained strengthening of the barrier. Further cytokine and ECIS studies showed that Imiquimod could abrogate some of the pro-inflammatory responses to Poly(I:C) and LPS. Most surprisingly, PCR revealed that the hCMVECs lacked TLR7 but expressed both TLR3 and TLR4 and did not respond to other structurally different TLR7 ligands. These data demonstrate that brain endothelial cells can be regulated by TLR 3 and TLR4 ligands in a pro-inflammatory manner and have receptors to Imiquimod, distinct to the classical TLR7, that function in an anti-inflammatory manner.
Collapse
Affiliation(s)
- Rebecca H Johnson
- Centre for Brain Research, Auckland, New Zealand.,Department of Pharmacology and Clinical Pharmacology, Auckland, New Zealand
| | - Dan T Kho
- Centre for Brain Research, Auckland, New Zealand.,Department of Pharmacology and Clinical Pharmacology, Auckland, New Zealand
| | - Simon J O' Carroll
- Centre for Brain Research, Auckland, New Zealand.,Department of Anatomy and Medical Imaging, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Catherine E Angel
- School of Biological Sciences, Faculty of Science, University of Auckland, Auckland, New Zealand
| | - E Scott Graham
- Centre for Brain Research, Auckland, New Zealand. .,Department of Pharmacology and Clinical Pharmacology, Auckland, New Zealand.
| |
Collapse
|
13
|
Wong TS, Chen S, Zhang MJ, Chan JYW, Gao W. Epstein-Barr virus-encoded microRNA BART7 downregulates major histocompatibility complex class I chain-related peptide A and reduces the cytotoxicity of natural killer cells to nasopharyngeal carcinoma. Oncol Lett 2018; 16:2887-2892. [PMID: 30127876 PMCID: PMC6096257 DOI: 10.3892/ol.2018.9041] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 04/26/2018] [Indexed: 01/10/2023] Open
Abstract
Evasion from natural killer (NK) cell surveillance enables cancer to proliferate and spread at the early stages. NK cells mediate specific cytolysis by activation of the triggering receptors on their cell surface, of which the communication between natural killer group 2, member D (NKG2D) and major histocompatibility complex class I chain-related peptide A (MICA) is a key regulatory axis. It has been indicated that cancer cells can reduce the surface expression of MICA, which thereby reduces the cytotoxicity of NK cells. In nasopharyngeal carcinoma (NPC), however, the underlying mechanism remains unclear. The present study indicated that MICA expression in NPC was regulated by TGFβ1. Furthermore, the human MICA gene was demonstrated to contain the c-Myc binding site in the promoter region. Notably, the results suggested that TGFβ1 upregulated MICA expression by promoting c-Myc expression. Additionally, the findings demosntrated that TGFβ1 expression in NPC was negatively controlled by Epstein-Barr virus-encoded microRNA BART7 (ebv-miR-BART7). In ebv-miR-BART7-expressing NPC, the TGFβ1/c-Myc/MICA regulatory axis was significantly inhibited. Notably, functional analysis indicated that NPC cells expressing ebv-miR-BART7 were less sensitive to the cytolysis mediated by NK cells. In conclusion, the present results revealed that ebv-miR-BART7-expressing NPC may impair NK cells recognition and activity, which suggests that targeting ebv-miR-BART7 may be a useful therapeutic strategy in NPC immunotherapy.
Collapse
Affiliation(s)
- Thian-Sze Wong
- Department of Surgery, The University of Hong Kong, Hong Kong SAR, P.R. China
| | - Siqi Chen
- Department of Surgery, The University of Hong Kong, Hong Kong SAR, P.R. China
| | - Min-Juan Zhang
- Department of Surgery, The University of Hong Kong, Hong Kong SAR, P.R. China
| | - Jimmy Yu-Wai Chan
- Department of Surgery, The University of Hong Kong, Hong Kong SAR, P.R. China
| | - Wei Gao
- Department of Surgery, The University of Hong Kong, Hong Kong SAR, P.R. China
| |
Collapse
|
14
|
Wang X, Liu A, Xing Y, Duan H, Xu W, Zhou Q, Wu H, Chen C, Chen B. Three-dimensional graphene biointerface with extremely high sensitivity to single cancer cell monitoring. Biosens Bioelectron 2018; 105:22-28. [DOI: 10.1016/j.bios.2018.01.012] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 01/02/2018] [Accepted: 01/08/2018] [Indexed: 10/18/2022]
|
15
|
Burmakina G, Bliznetsov K, Malogolovkin A. Real-time analysis of the cytopathic effect of African swine fever virus. J Virol Methods 2018; 257:58-61. [PMID: 29627336 DOI: 10.1016/j.jviromet.2018.04.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 03/19/2018] [Accepted: 04/04/2018] [Indexed: 11/28/2022]
Abstract
Conventional methods, which quantitatively assess virus replication, are based on direct examination of viral cytopathic effect (CPE), which is time consuming, tedious and based on endpoint reading. The Real-Time Cell Analysis (RTCA) xCELLigence® system offers an alternative approach to evaluate virus-induced CPE, and here was evaluated as a means to dynamically assess CPE caused by African swine fever virus (ASFV). RTCA was used to identify optimum time for ASFV infection based on cell index (CI) and to evaluate ASFV CPE kinetics in COS-1 cells. Data indicated that the RTCA has tremendous potential to methodologically and quantitatively improve assays used to study efficiency of ASFV drug inhibitors and neutralizing antibodies.
Collapse
Affiliation(s)
- Galina Burmakina
- Federal Research Center for Virology and Microbiology (FRCVM), 601125, Volginskiy, Bakulova 1, Russia
| | | | - Alexander Malogolovkin
- Federal Research Center for Virology and Microbiology (FRCVM), 601125, Volginskiy, Bakulova 1, Russia.
| |
Collapse
|
16
|
Abstract
The use of impedance-based label free cell analysis is increasingly popular and has many different applications. Here, we report that a real-time cell analyzer (RTCA) can be used to study the stimulation of Natural Killer (NK) cells. Engagement of NK cells via plate-bound antibodies directed against different activating surface receptors could be measured in real time using the label-free detection of impedance. The change in impedance was dependent on early signal transduction events in the NK cells as it was blocked by inhibitors of Src-family kinases and by inhibiting actin polymerization. While CD16 was the only receptor that could induce a strong change in impedance in primary NK cells, several activating receptors induced changes in impedance in expanded NK cells. Using PBMCs we could detect T cell receptor-mediated T cell activation and CD16-mediated NK cell activation in the same sample. Performing a dose-response analysis for the Src-family kinases inhibitor PP1 we show that T cells are more sensitive to inhibition compared to NK cells. Our data demonstrate that the RTCA can be used to detect physiological activation events in NK cells in a label-free and real-time fashion.
Collapse
|
17
|
Xu Z, Song Y, Jiang H, Kong Y, Li X, Chen J, Wu Y. Regeneration of Arrayed Gold Microelectrodes Equipped for a Real-Time Cell Analyzer. J Vis Exp 2018. [PMID: 29578508 DOI: 10.3791/56250] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The label-free cell-based assay is advantageous for biochemical study because of it does not require the use of experimental animals. Due to its ability to provide more dynamic information about cells under physiological conditions than classical biochemical assays, this label-free real-time cell assay based on the electric impedance principle is attracting more attention during the past decade. However, its practical utilization may be limited due to the relatively expensive cost of measurement, in which costly consumable disposable gold microchips are used for the cell analyzer. In this protocol, we have developed a general strategy to regenerate arrayed gold microelectrodes equipped for a commercial label-free cell analyzer. The regeneration process includes trypsin digestion, rinsing with ethanol and water, and a spinning step. The proposed method has been tested and shown to be effective for the regeneration and repeated usage of commercial electronic plates at least three times, which will help researchers save on the high running cost of real-time cell assays.
Collapse
Affiliation(s)
- Zhihui Xu
- School of Public Health, Nanjing Medical University
| | - Yiyan Song
- School of Public Health, Nanjing Medical University
| | | | - Yan Kong
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Chemistry and Chemical Engineering, Nanjing Tech University
| | - Xiaoming Li
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Chemistry and Chemical Engineering, Nanjing Tech University
| | - Jin Chen
- School of Public Health, Nanjing Medical University;
| | - Yuan Wu
- Department of Medical Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University;
| |
Collapse
|
18
|
Bowen KE, Mathew SO, Borgmann K, Ghorpade A, Mathew PA. A novel ligand on astrocytes interacts with natural cytotoxicity receptor NKp44 regulating immune response mediated by NK cells. PLoS One 2018; 13:e0193008. [PMID: 29447242 PMCID: PMC5814005 DOI: 10.1371/journal.pone.0193008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 01/16/2018] [Indexed: 12/29/2022] Open
Abstract
NK cells play important role in immunity against pathogens and cancer. NK cell functions are regulated by inhibitory and activating receptors binding corresponding ligands on the surface of target cells. NK cells were shown to be recruited to the CNS following several pathological conditions. NK cells could impact CNS physiology by killing glial cells and by secreting IFN-γ. Astrocytes are intimately involved in immunological and inflammatory events occurring in the CNS and reactive astrogliosis is a key feature in HIV-associated neurocognitive disorders. There is little data on NK-astrocyte interactions and ligands expressed on astrocytes that could impact NK cell function. Natural cytotoxicity receptors (NCRs) play a critical role in the cytolytic function of NK cells. Among the NCRs, NKp44 is unique in expression and signal transduction. NKp44 is expressed only upon activation of NK cells and it can mediate both activating and inhibitory signals to NK cells. Here, we have studied the expression and function of natural cytotoxicity receptor NKp44 upon NK-astrocytes interactions in the presence or absence of an HIV peptide (HIV-3S peptide) shown to induce NK cell killing of CD4+ T cells during HIV–infection. Using a fusion protein consisting of the extracellular domain of NKp44 fused to Fc portion of human IgG, we determined the expression of a novel ligand for NKp44 (NKp44L) on astrocytes. Incubation of astrocytes with HIV-3S peptide downregulated NKp44L expression on astrocytes implicating protection from NK mediated killing. Thus, our study showed that NKp44 have a protective effect on astrocytes from NK cell mediated killing during HIV infection and impact astrocyte role in HAND.
Collapse
Affiliation(s)
- Kelly E Bowen
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
| | - Stephen O Mathew
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
| | - Kathleen Borgmann
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
| | - Anuja Ghorpade
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
| | - Porunelloor A Mathew
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
| |
Collapse
|
19
|
Türker Şener L, Albeniz G, Dinç B, Albeniz I. iCELLigence real-time cell analysis system for examining the cytotoxicity of drugs to cancer cell lines. Exp Ther Med 2017; 14:1866-1870. [PMID: 28962095 PMCID: PMC5609197 DOI: 10.3892/etm.2017.4781] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 03/23/2017] [Indexed: 12/20/2022] Open
Abstract
The recently developed iCELLigence™ real-time cell analyzer (RTCA) can be used for the label-free real-time monitoring of cancer cell proliferation, viability, invasion and cytotoxicity. The RTCA system uses 16-well microtiter plates with a gold microelectrode biosensor array that measures impedance when cells adhere to the microelectrodes causing an alternating current. By measuring the electric field generated in this process, the RTCA system can be used for the analysis of cell proliferation, viability, morphology and migration. The present review aimed to summarize the working method of the RTCA system, in addition to discussing the research performed using the system for various applications, including cancer drug discovery via measuring cytotoxicity.
Collapse
Affiliation(s)
- Leyla Türker Şener
- Department of Biophysics, Faculty of Medicine, Istanbul University, 34093 Istanbul, Turkey
| | - Gürcan Albeniz
- Department of General Surgery, Cerrahpaşa Faculty of Medicine, Istanbul University, 34096 Istanbul, Turkey
| | - Bircan Dinç
- Department of Basic Sciences, School of Engineering and Architecture, Istanbul Kemerburgaz University, 34394 Istanbul, Turkey
| | - Işil Albeniz
- Department of Biophysics, Faculty of Medicine, Istanbul University, 34093 Istanbul, Turkey
| |
Collapse
|
20
|
Yang M, Liu Y, Hou W, Zhi X, Zhang C, Jiang X, Pan F, Yang Y, Ni J, Cui D. Mitomycin C-treated human-induced pluripotent stem cells as a safe delivery system of gold nanorods for targeted photothermal therapy of gastric cancer. NANOSCALE 2017; 9:334-340. [PMID: 27922138 DOI: 10.1039/c6nr06851k] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Human-induced pluripotent stem cells (iPS) possess an intrinsic tumor tropism ability. However, iPS cells are impeded in clinical applications of tumor therapy due to the formation of teratomas and their survival in normal organs such as the liver, lungs, spleen and kidneys. Mitomycin C (MMC) can overcome this limitation by suppressing iPS proliferation. Herein, we fabricated a safe delivery system of iPS cells treated with MMC loading with gold nanorods (AuNRs) for the targeted photothermal treatment of gastric cancer. Our results showed that the tumor cells were efficiently killed by the heat generated from the gold nanorods, and the iPS cells ultimately died due to the action of MMC seven days after the photothermal treatment. This suggested that pre-treated iPS cells with MMC can be used as a novel and safe approach for targeted tumor therapy. This paves the road for clinical translation in the future.
Collapse
Affiliation(s)
- Meng Yang
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Centre for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
A general method to regenerate arrayed gold microelectrodes for label-free cell assay. Anal Biochem 2016; 516:57-60. [PMID: 27746100 DOI: 10.1016/j.ab.2016.10.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 10/10/2016] [Accepted: 10/13/2016] [Indexed: 12/29/2022]
Abstract
We developed a method to regenerate arrayed gold microelectrodes equipped for a commercial label-free cell analyzer. The regeneration process includes efficient treatment of the gold surface with trypsin (0.25%, v/v) digestion, rinsing with ethanol and deionized water and spinning steps. The proposed method ensured complete regeneration and repeated usage of gold microchips up to 4 times for the real-time electric impedance measurement of anti-cancer drug cytotoxicity.
Collapse
|
22
|
Collins AR, Annangi B, Rubio L, Marcos R, Dorn M, Merker C, Estrela-Lopis I, Cimpan MR, Ibrahim M, Cimpan E, Ostermann M, Sauter A, Yamani NE, Shaposhnikov S, Chevillard S, Paget V, Grall R, Delic J, de-Cerio FG, Suarez-Merino B, Fessard V, Hogeveen KN, Fjellsbø LM, Pran ER, Brzicova T, Topinka J, Silva MJ, Leite PE, Ribeiro AR, Granjeiro JM, Grafström R, Prina-Mello A, Dusinska M. High throughput toxicity screening and intracellular detection of nanomaterials. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2016; 9. [PMID: 27273980 PMCID: PMC5215403 DOI: 10.1002/wnan.1413] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 04/08/2016] [Accepted: 04/12/2016] [Indexed: 12/25/2022]
Abstract
With the growing numbers of nanomaterials (NMs), there is a great demand for rapid and reliable ways of testing NM safety—preferably using in vitro approaches, to avoid the ethical dilemmas associated with animal research. Data are needed for developing intelligent testing strategies for risk assessment of NMs, based on grouping and read‐across approaches. The adoption of high throughput screening (HTS) and high content analysis (HCA) for NM toxicity testing allows the testing of numerous materials at different concentrations and on different types of cells, reduces the effect of inter‐experimental variation, and makes substantial savings in time and cost. HTS/HCA approaches facilitate the classification of key biological indicators of NM‐cell interactions. Validation of in vitroHTS tests is required, taking account of relevance to in vivo results. HTS/HCA approaches are needed to assess dose‐ and time‐dependent toxicity, allowing prediction of in vivo adverse effects. Several HTS/HCA methods are being validated and applied for NM testing in the FP7 project NANoREG, including Label‐free cellular screening of NM uptake, HCA, High throughput flow cytometry, Impedance‐based monitoring, Multiplex analysis of secreted products, and genotoxicity methods—namely High throughput comet assay, High throughput in vitro micronucleus assay, and γH2AX assay. There are several technical challenges with HTS/HCA for NM testing, as toxicity screening needs to be coupled with characterization of NMs in exposure medium prior to the test; possible interference of NMs with HTS/HCA techniques is another concern. Advantages and challenges of HTS/HCA approaches in NM safety are discussed. WIREs Nanomed Nanobiotechnol 2017, 9:e1413. doi: 10.1002/wnan.1413 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Andrew R Collins
- Comet Biotech AS, and Department of Nutrition, University of Oslo, Norway
| | | | - Laura Rubio
- Grup de Mutagènesi, Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Ricard Marcos
- Grup de Mutagènesi, Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Spain.,CIBER Epidemiología y Salud Pública, ISCIII, Spain
| | - Marco Dorn
- Institute of Biophysics and Medical Physics, University of Leipzig, Leipzig, Germany
| | - Carolin Merker
- Institute of Biophysics and Medical Physics, University of Leipzig, Leipzig, Germany
| | - Irina Estrela-Lopis
- Institute of Biophysics and Medical Physics, University of Leipzig, Leipzig, Germany
| | - Mihaela Roxana Cimpan
- Department of Clinical Dentistry, Faculty of Medicine and Dentistry, University of Bergen, Norway
| | - Mohamed Ibrahim
- Department of Clinical Dentistry, Faculty of Medicine and Dentistry, University of Bergen, Norway
| | - Emil Cimpan
- Department of Electrical Engineering, Faculty of Engineering, Bergen University College, Norway
| | - Melanie Ostermann
- Department of Clinical Dentistry, Faculty of Medicine and Dentistry, University of Bergen, Norway
| | - Alexander Sauter
- Department of Clinical Dentistry, Faculty of Medicine and Dentistry, University of Bergen, Norway
| | - Naouale El Yamani
- Comet Biotech AS, and Department of Nutrition, University of Oslo, Norway.,Health Effects Group, Department of Environmental Chemistry, NILU- Norwegian Institute for Air Research, Kjeller, Norway
| | | | - Sylvie Chevillard
- Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA) Direction des Sciences du Vivant, Institut de Radiobiologie Cellulaire et Moléculaire, Service de Radiobiologie Expérimentale et d'Innovation Technologique, Laboratoire de Cancérologie Expérimentale, Fontenay-aux-Roses cedex, France
| | - Vincent Paget
- Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA) Direction des Sciences du Vivant, Institut de Radiobiologie Cellulaire et Moléculaire, Service de Radiobiologie Expérimentale et d'Innovation Technologique, Laboratoire de Cancérologie Expérimentale, Fontenay-aux-Roses cedex, France
| | - Romain Grall
- Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA) Direction des Sciences du Vivant, Institut de Radiobiologie Cellulaire et Moléculaire, Service de Radiobiologie Expérimentale et d'Innovation Technologique, Laboratoire de Cancérologie Expérimentale, Fontenay-aux-Roses cedex, France
| | - Jozo Delic
- Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA) Direction des Sciences du Vivant, Institut de Radiobiologie Cellulaire et Moléculaire, Service de Radiobiologie Expérimentale et d'Innovation Technologique, Laboratoire de Cancérologie Expérimentale, Fontenay-aux-Roses cedex, France
| | | | | | - Valérie Fessard
- ANSES Fougères Laboratory, Contaminant Toxicology Unit, France
| | | | - Lise Maria Fjellsbø
- Health Effects Group, Department of Environmental Chemistry, NILU- Norwegian Institute for Air Research, Kjeller, Norway
| | - Elise Runden Pran
- Health Effects Group, Department of Environmental Chemistry, NILU- Norwegian Institute for Air Research, Kjeller, Norway
| | - Tana Brzicova
- Institute of Experimental Medicine AS CR, Prague, Czech Republic
| | - Jan Topinka
- Institute of Experimental Medicine AS CR, Prague, Czech Republic
| | - Maria João Silva
- Human Genetics Department, National Institute of Health Doutor Ricardo Jorge and Centre for Toxicogenomics and Human Health, NMS/FCM, UNL, Lisbon, Portugal
| | - P E Leite
- Directory of Life Sciences Applied Metrology, National Institute of Metrology Quality and Technology, Rio de Janeiro, Brazil
| | - A R Ribeiro
- Directory of Life Sciences Applied Metrology, National Institute of Metrology Quality and Technology, Rio de Janeiro, Brazil
| | - J M Granjeiro
- Directory of Life Sciences Applied Metrology, National Institute of Metrology Quality and Technology, Rio de Janeiro, Brazil
| | - Roland Grafström
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Adriele Prina-Mello
- Nanomedicine Group, Trinity Centre for Health Sciences, Trinity College Dublin, Dublin, Ireland
| | - Maria Dusinska
- Health Effects Group, Department of Environmental Chemistry, NILU- Norwegian Institute for Air Research, Kjeller, Norway
| |
Collapse
|
23
|
Xu Y, Xie X, Duan Y, Wang L, Cheng Z, Cheng J. A review of impedance measurements of whole cells. Biosens Bioelectron 2016; 77:824-36. [DOI: 10.1016/j.bios.2015.10.027] [Citation(s) in RCA: 252] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Revised: 10/03/2015] [Accepted: 10/09/2015] [Indexed: 11/17/2022]
|
24
|
Regulation of human cerebro-microvascular endothelial baso-lateral adhesion and barrier function by S1P through dual involvement of S1P1 and S1P2 receptors. Sci Rep 2016; 6:19814. [PMID: 26813587 PMCID: PMC4728386 DOI: 10.1038/srep19814] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 11/30/2015] [Indexed: 01/07/2023] Open
Abstract
Herein we show that S1P rapidly and acutely reduces the focal adhesion strength and barrier tightness of brain endothelial cells. xCELLigence biosensor technology was used to measure focal adhesion, which was reduced by S1P acutely and this response was mediated through both S1P1 and S1P2 receptors. S1P increased secretion of several pro-inflammatory mediators from brain endothelial cells. However, the magnitude of this response was small in comparison to that mediated by TNFα or IL-1β. Furthermore, S1P did not significantly increase cell-surface expression of any key cell adhesion molecules involved in leukocyte recruitment, included ICAM-1 and VCAM-1. Finally, we reveal that S1P acutely and dynamically regulates microvascular endothelial barrier tightness in a manner consistent with regulated rapid opening followed by closing and strengthening of the barrier. We hypothesise that the role of the S1P receptors in this process is not to cause barrier dysfunction, but is related to controlled opening of the endothelial junctions. This was revealed using real-time measurement of barrier integrity using ECIS ZΘ TEER technology and endothelial viability using xCELLigence technology. Finally, we show that these responses do not occur simply though the pharmacology of a single S1P receptor but involves coordinated action of S1P1 and S1P2 receptors.
Collapse
|
25
|
Mustafa AM, Caprioli G, Dikmen M, Kaya E, Maggi F, Sagratini G, Vittori S, Öztürk Y. Evaluation of neuritogenic activity of cultivated, wild and commercial roots of Gentiana lutea L. J Funct Foods 2015. [DOI: 10.1016/j.jff.2015.09.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
26
|
Ryan CM, Brown JAL, Bourke E, Prendergast ÁM, Kavanagh C, Liu Z, Owens P, Shaw G, Kolch W, O'Brien T, Barry FP. ROCK activity and the Gβγ complex mediate chemotactic migration of mouse bone marrow-derived stromal cells. Stem Cell Res Ther 2015. [PMID: 26204937 PMCID: PMC4603944 DOI: 10.1186/s13287-015-0125-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Bone marrow-derived stromal cells (BMSCs), also known as mesenchymal stem cells, are the focus of intensive efforts worldwide to elucidate their function and biology. Despite the importance of BMSC migration for their potential therapeutic uses, the mechanisms and signalling governing stem cell migration are still not fully elucidated. METHODS We investigated and detailed the effects of MCP-1 activation on BMSCs by using inhibitors of G protein-coupled receptor alpha beta (GPCR αβ), ROCK (Rho-associated, coiled-coil containing protein kinase), and PI3 kinase (PI3K). The effects of MCP-1 stimulation on intracellular signalling cascades were characterised by using immunoblotting and immunofluorescence. The effectors of MCP-1-mediated migration were investigated by using migration assays (both two-dimensional and three-dimensional) in combination with inhibitors. RESULTS We established the kinetics of the MCP-1-activated signalling cascade and show that this cascade correlates with cell surface re-localisation of chemokine (C motif) receptor 2 (CCR2) (the MCP-1 receptor) to the cell periphery following MCP-1 stimulation. We show that MCP-1-initiated signalling is dependent on the activation of βγ subunits from the GPCR αβγ complex. In addition, we characterise a novel role for PI3Kγ signalling for the activation of both PAK and ERK following MCP-1 stimulation. We present evidence that the Gβγ complex is responsible for PI3K/Akt, PAK, and ERK signalling induced by MCP-1 in BMSCs. Importantly, we found that, in BMSCs, inhibition of ROCK significantly inhibits MCP-1-induced chemotactic migration, in contrast to previous reports in other systems. CONCLUSIONS Our results indicate differential chemotactic signalling in mouse BMSCs, which has important implications for the translation of in vivo mouse model findings into human trials. We identified novel components and interactions activated by MCP-1-mediated signalling, which are important for stem cell migration. This work has identified additional potential therapeutic targets that could be manipulated to improve BMSC delivery and homing.
Collapse
Affiliation(s)
- Caroline M Ryan
- Regenerative Medicine Institute (REMEDI), Biosciences, National University of Ireland Galway, University Road, Galway, Ireland. .,Systems Biology Ireland, UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland.
| | - James A L Brown
- Regenerative Medicine Institute (REMEDI), Biosciences, National University of Ireland Galway, University Road, Galway, Ireland. .,Systems Biology Ireland, UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland. .,Present address: Discipline of Surgery, School of Medicine, Lambe Institute, Translational Research Facility, National University of Ireland Galway, University Road, Galway, Ireland.
| | - Emer Bourke
- Discipline of Pathology, School of Medicine, National University of Ireland Galway, University Road, Galway, Ireland.
| | - Áine M Prendergast
- Regenerative Medicine Institute (REMEDI), Biosciences, National University of Ireland Galway, University Road, Galway, Ireland. .,Systems Biology Ireland, UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland. .,Present address: Hematopoietic Stem Cells and Stress' group, Division of Stem Cells and Cancer, Deutsches Krebsforschungszentrum (DKFZ), Im Neuenheimer feld 280, 69120, Heidelberg, Germany.
| | - Claire Kavanagh
- Regenerative Medicine Institute (REMEDI), Biosciences, National University of Ireland Galway, University Road, Galway, Ireland.
| | - Zhonglin Liu
- Department of Radiology, University of Arizona, Tucson, AZ, 85724-5067, USA.
| | - Peter Owens
- Centre for Micro and Imaging, National University of Ireland Galway, University Road, Galway, Ireland.
| | - Georgina Shaw
- Regenerative Medicine Institute (REMEDI), Biosciences, National University of Ireland Galway, University Road, Galway, Ireland.
| | - Walter Kolch
- Systems Biology Ireland, UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Timothy O'Brien
- Regenerative Medicine Institute (REMEDI), Biosciences, National University of Ireland Galway, University Road, Galway, Ireland. .,Systems Biology Ireland, UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Frank P Barry
- Regenerative Medicine Institute (REMEDI), Biosciences, National University of Ireland Galway, University Road, Galway, Ireland. .,Systems Biology Ireland, UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
27
|
Mustafa AM, Maggi F, Papa F, Kaya E, Dikmen M, Öztürk Y. Isofuranodiene: A neuritogenic compound isolated from wild celery (Smyrnium olusatrum L., Apiaceae). Food Chem 2015; 192:782-7. [PMID: 26304411 DOI: 10.1016/j.foodchem.2015.07.079] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 07/04/2015] [Accepted: 07/20/2015] [Indexed: 12/24/2022]
Abstract
In the search for neuroactive compounds that mimic the nerve growth factor (NGF) activity for the protection against neurodegenerative diseases, the potential medicinal values of foods and plants attracts intense interest. Isofuranodiene is the major constituent of the essential oil of wild celery (Smyrnium olusatrum L., Apiaceae). The cytotoxic effects of isofuranodiene towards rat neuronal PC-12 pheochromocytoma cells were determined by MTT assay, while the cell differentiation was evaluated with xCELLigence real time cell analysis system (RTCA DP), and the neuritogenic activity was assessed by neurite outgrowth image analysis. Isofuranodiene at concentrations of 25 and 12.5 μM alone, or in combination with 50 nM NGF, showed a marked stimulation of neuritogenesis, but it was more effective at 12.5 μM with or without NGF. The present study reports the first evidence of the neuritogenic effects of isofuranodiene, which appears to be a promising neurotrophic and neuroprotective agent deserving further investigation.
Collapse
Affiliation(s)
- Ahmed M Mustafa
- School of Pharmacy, University of Camerino, Via Sant'Agostino 1, 62032 Camerino, Italy
| | - Filippo Maggi
- School of Pharmacy, University of Camerino, Via Sant'Agostino 1, 62032 Camerino, Italy.
| | - Fabrizio Papa
- School of Science and Technology, University of Camerino, Via Sant'Agostino 1, 62032 Camerino, Italy
| | - Elif Kaya
- Department of Pharmacology, Faculty of Pharmacy, Anadolu University, 26470 Eskisehir, Turkey
| | - Miris Dikmen
- Department of Pharmacology, Faculty of Pharmacy, Anadolu University, 26470 Eskisehir, Turkey
| | - Yusuf Öztürk
- Department of Pharmacology, Faculty of Pharmacy, Anadolu University, 26470 Eskisehir, Turkey
| |
Collapse
|
28
|
Application of xCELLigence RTCA Biosensor Technology for Revealing the Profile and Window of Drug Responsiveness in Real Time. BIOSENSORS-BASEL 2015; 5:199-222. [PMID: 25893878 PMCID: PMC4493546 DOI: 10.3390/bios5020199] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 03/27/2015] [Accepted: 04/02/2015] [Indexed: 12/12/2022]
Abstract
The xCELLigence technology is a real-time cellular biosensor, which measures the net adhesion of cells to high-density gold electrode arrays printed on custom-designed E-plates. The strength of cellular adhesion is influenced by a myriad of factors that include cell type, cell viability, growth, migration, spreading and proliferation. We therefore hypothesised that xCELLigence biosensor technology would provide a valuable platform for the measurement of drug responses in a multitude of different experimental, clinical or pharmacological contexts. In this manuscript, we demonstrate how xCELLigence technology has been invaluable in the identification of (1) not only if cells respond to a particular drug, but (2) the window of drug responsiveness. The latter aspect is often left to educated guess work in classical end-point assays, whereas biosensor technology reveals the temporal profile of the response in real time, which enables both acute responses and longer term responses to be profiled within the same assay. In our experience, the xCELLigence biosensor technology is suitable for highly targeted drug assessment and also low to medium throughput drug screening, which produces high content temporal data in real time.
Collapse
|
29
|
Laird D, El-Baba MD, Charpin-El Hamri G, Eberwein P, Nelson K, Tomakidi P, Steinberg T. In vitro and in vivo biocompatibility evaluation of a novobiocin stimulus-responsive poly(ethylene glycol)-based hydrogel designed for soft tissue regeneration. J BIOACT COMPAT POL 2015. [DOI: 10.1177/0883911515570370] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
In vitro and in vivo biocompatibility evaluation of medical devices designed for soft tissue healing and regeneration is mandatory for new biomaterials targeted toward human application. To this end, a novel stimulus-responsive eight-arm poly(ethylene glycol) (PEG)-based hydrogel for soft tissue regeneration was subjected to preclinical safety evaluation. While in vitro safety testing included determination of half-maximal inhibitory concentrations by cell index impedance measurements, an in vivo pilot rat study focused on detectable signs of oral mucosal inflammation. In a hydrogel with a total volume of 40 µL, the following concentrations of the constituents were released upon hydrogel dissolution in 1 mL body fluid: PEG, 19 µM; gyrase B, 152 µM; and coumermycin, 76 µM, while the dissolving agent novobiocin was effective in a range from 50 to 100 µM. Concerning the components’ biological effects, half-maximal inhibitory concentrations of 806 and 95 µM for human oral mucosal gingival fibroblasts and gingival keratinocytes were calculated for novobiocin, proving gingival keratinocytes more susceptible to substance effects. Furthermore, with respect to coumermycin concentrations, half-maximal inhibitory concentrations of 10 and 3 µM for gingival fibroblasts and gingival keratinocytes, respectively, were obtained, and thereby revealed coumermycin to be toxic in vitro. In vivo, submucosal hard palatal implantation of standard hydrogels showed no evidence of inflammation at any time point under study, which was supported by immunohistochemistry analysis. In order to realize the potential of such a promising stimulus-responsive hydrogel biomaterial concept for soft tissue wound healing and/or regenerative applications, the crosslinker coumermycin and its competitor novobiocin should be replaced by more innocuous alternative protein–ligand pairs.
Collapse
Affiliation(s)
- Dougal Laird
- Department of Oral Biotechnology, University Medical Center Freiburg, Freiburg, Germany
| | | | | | - Philipp Eberwein
- Department of Ophthalmology, University Eye Hospital Freiburg, Freiburg, Germany
| | - Katja Nelson
- Department of Craniomaxillofacial Surgery, University Medical Center Freiburg, Freiburg, Germany
| | - Pascal Tomakidi
- Department of Oral Biotechnology, University Medical Center Freiburg, Freiburg, Germany
| | - Thorsten Steinberg
- Department of Oral Biotechnology, University Medical Center Freiburg, Freiburg, Germany
| |
Collapse
|
30
|
Pham PV, Nguyen NT, Nguyen HM, Khuat LT, Le PM, Pham VQ, Nguyen ST, Phan NK. A simple in vitro method for evaluating dendritic cell-based vaccinations. Onco Targets Ther 2014; 7:1455-64. [PMID: 25170272 PMCID: PMC4145728 DOI: 10.2147/ott.s67057] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Background Dendritic cell (DC) therapy is a promising therapy for cancer-targeting treatments. Recently, DCs have been used for treatment of some cancers. We aimed to develop an in vitro assay to evaluate DC therapy in cancer treatment using a breast cancer model. Methods DCs were induced from murine bone marrow mononuclear cells in Roswell Park Memorial Institute (RPMI) 1640 medium supplemented with GM-CSF (20 ng/mL) and IL-4 (20 ng/mL). Immature DCs were primed with breast cancer stem cell (BCSC)-derived antigens. BCSCs were sorted from 4T1 cell lines based on aldehyde dehydrogenase expression. A mixture of DCs and cytotoxic T lymphocytes (CTLs) were used to evaluate the inhibitory effect of antigen-primed DCs on BCSCs. BCSC proliferation and doubling time were recorded based on impedance-based cell analysis using the xCELLigence system. The specification of inhibitory effects of DCs and CTLs was also evaluated using the same system. Results The results showed that impedance-based analysis of BCSCs reflected cytotoxicity and inhibitory effects of DCs and CTLs at 72 hours. Differences in ratios of DC:CTL changed the cytotoxicity of DCs and CTLs. Conclusion This study successfully used impedance-based cell analysis as a new in vitro assay to evaluate DC efficacy in cancer immunotherapy. We hope this technique will contribute to the development and improvement of immunotherapies in the near future.
Collapse
Affiliation(s)
- Phuc Van Pham
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam
| | - Nhung Thi Nguyen
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam
| | - Hoang Minh Nguyen
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam
| | - Lam Tan Khuat
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam
| | - Phong Minh Le
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam
| | - Viet Quoc Pham
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam
| | - Sinh Truong Nguyen
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam
| | - Ngoc Kim Phan
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam
| |
Collapse
|
31
|
MacDonald C, Unsworth CP, Graham ES. Enrichment of differentiated hNT neurons and subsequent analysis using flow-cytometry and xCELLigence sensing. J Neurosci Methods 2014; 227:47-56. [PMID: 24530700 DOI: 10.1016/j.jneumeth.2014.02.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Revised: 02/02/2014] [Accepted: 02/04/2014] [Indexed: 12/22/2022]
Abstract
BACKGROUND Human neurons (hNT neurons), obtained from the NTera2/D1 precursor cell line, are highly valued by many neuroscientists as isolation of adult human primary neuronal cells continues to elude us. hNT neurons are generated by differentiation of the NT2 precursors for a period of 4 weeks followed by 2 weeks of mitotic inhibition. This yields a heterogeneous population of neuronal phenotypes and underlying astrocyte precursors, the latter of which are very difficult to visualise using standard light microscopy. Such a mixed culture is acceptable for some applications (e.g. measurement of synaptic plasticity), whereas others (e.g. proteomics or transcriptomics) require almost pure cultures of hNT neurons. NEW METHOD Here we describe a simple method for obtaining highly enriched cultures of hNT neurons following the first neuronal harvest and detail several additional methods, namely flow-cytometry and xCELLigence© biosensor technology, to rapidly and reliably determine the purity and viability of the cultures. COMPARISON WITH EXISTING METHODS This method of enrichment for the neurons is novel and advances the end user applications of the cells. RESULTS In addition, we apply the enrichment method to conduct analysis of cell-surface markers using flow-cytometry on the enriched neuronal cells. Furthermore, we apply this method to generate enriched neuronal cells on which we conduct analysis of cell-surface markers using flow-cytometry. CONCLUSIONS Collectively, this paper describes several new advances, which will create opportunities when using these cells and similar preparations, and provides the protocol for analysis of these cells using flow-cytometry and biosensor technology.
Collapse
Affiliation(s)
- Christa MacDonald
- Department of Pharmacology and Centre for Brain Research, School of Medical Sciences, The University of Auckland, Faculty of Medical and Health Sciences, Private Bag 92019, Auckland 1142, New Zealand; Department of Engineering Science, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Charles P Unsworth
- Department of Engineering Science, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - E Scott Graham
- Department of Pharmacology and Centre for Brain Research, School of Medical Sciences, The University of Auckland, Faculty of Medical and Health Sciences, Private Bag 92019, Auckland 1142, New Zealand.
| |
Collapse
|
32
|
Kim JH, Jekarl DW, Kim M, Oh EJ, Kim Y, Park IY, Shin JC. Effects of ECM protein mimetics on adhesion and proliferation of chorion derived mesenchymal stem cells. Int J Med Sci 2014; 11:298-308. [PMID: 24516355 PMCID: PMC3917120 DOI: 10.7150/ijms.6672] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2013] [Accepted: 08/12/2013] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND We evaluated the effects of fibronectin, collagen, cadherin, and laminin based extracellular matrix (ECM) protein mimetics coated with mussel derived adhesive protein (MAP) on adhesion and proliferation of chorionic mesenchymal stem cells (cMSCs). METHODS Human placental chorionic tissues from term third-trimester pregnancies (n=3) were used. The cMSCs were cultured on rationally designed ECM protein mimetics coated with MAP on plastic surfaces with the addition of reduced fetal bovine serum (0.5%, 1% FBS). Adhesion capabilities were monitored by a real time cell analysis system (RTCA) utilizing an impedance method. Proliferation capabilities were monitored by RTCA and MTS assay. RESULTS Of the ECM protein mimetics tested, GRGDSP(FN) coated surfaces exhibited the highest adhesion and proliferation capabilities on RTCA at FBS concentration of 0.5% and 1%. When 0.5% FBS was added to ECM protein mimetics during the MTS assay, GRGDSP(FN), REDV(FN), and collagen mimetics, GPKGAAGEPGKP(ColI) showed higher cMSCs proliferation compared with the control. When 1% FBS was added, GRGDSP(FN) and TAIPSCPEGTVPLYS(ColIV) showed significant cMSCs proliferation capacity. CONCLUSIONS Fibronectin mimetics, GRGDSP(FN) amino acid sequence showed the highest adhesion and proliferation capabilities. In addition, results from RTCA assessment of cell viability correlated well with the tetrazolium-based MTS assay.
Collapse
Affiliation(s)
- Ji-Hyun Kim
- 1. Department of Laboratory Medicine College of Medicine, The Catholic University of Korea, Seoul, South Korea; ; 3. Department of Biomedical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Dong Wook Jekarl
- 1. Department of Laboratory Medicine College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Myungshin Kim
- 1. Department of Laboratory Medicine College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Eun-Jee Oh
- 1. Department of Laboratory Medicine College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Yonggoo Kim
- 1. Department of Laboratory Medicine College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - In Yang Park
- 2. Department of Obstetrics and Gynecology, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Jong Chul Shin
- 2. Department of Obstetrics and Gynecology, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
33
|
Exposure to inflammatory cytokines IL-1β and TNFα induces compromise and death of astrocytes; implications for chronic neuroinflammation. PLoS One 2013; 8:e84269. [PMID: 24367648 PMCID: PMC3868583 DOI: 10.1371/journal.pone.0084269] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 11/21/2013] [Indexed: 02/06/2023] Open
Abstract
Background Astrocytes have critical roles in the human CNS in health and disease. They provide trophic support to neurons and are innate-immune cells with keys roles during states-of-inflammation. In addition, they have integral functions associated with maintaining the integrity of the blood-brain barrier. Methods We have used cytometric bead arrays and xCELLigence technology to monitor the to monitor the inflammatory response profiles and astrocyte compromise in real-time under various inflammatory conditions. Responses were compared to a variety of inflammatory cytokines known to be released in the CNS during neuroinflammation. Astrocyte compromise measured by xCELLigence was confirmed using ATP measurements, cleaved caspase 3 expression, assessment of nuclear morphology and cell death. Results Inflammatory activation (IL-1β or TNFα) of astrocytes results in the transient production of key inflammatory mediators including IL-6, cell surface adhesion molecules, and various leukocyte chemoattractants. Following this phase, the NT2-astrocytes progressively become compromised, which is indicated by a loss of adhesion, appearance of apoptotic nuclei and reduction in ATP levels, followed by DEATH. The earliest signs of astrocyte compromise were observed between 24-48h post cytokine treatment. However, significant cell loss was not observed until at least 72h, where there was also an increase in the expression of cleaved-caspase 3. By 96 hours approximately 50% of the astrocytes were dead, with many of the remaining showing signs of compromise too. Numerous other inflammatory factors were tested, however these effects were only observed with IL-1β or TNFα treatment. Conclusions Here we reveal direct sensitivity to mediators of the inflammatory milieu. We highlight the power of xCELLigence technology for revealing the early progressive compromise of the astrocytes, which occurs 24-48 hours prior to substantive cell loss. Death induced by IL-1β or TNFα is relevant clinically as these two cytokines are produced by various peripheral tissues and by resident brain cells.
Collapse
|
34
|
Park KH, Park H, Kim M, Kim Y, Han K, Oh EJ. Evaluation of NK cell function by flowcytometric measurement and impedance based assay using real-time cell electronic sensing system. BIOMED RESEARCH INTERNATIONAL 2013; 2013:210726. [PMID: 24236291 PMCID: PMC3819884 DOI: 10.1155/2013/210726] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Revised: 08/22/2013] [Accepted: 09/06/2013] [Indexed: 12/13/2022]
Abstract
Although real-time cell electronic sensing (RT-CES) system-based natural killer (NK) cytotoxicity has been introduced, it has not been evaluated using human blood samples. In present study, we measured flowcytometry based assay (FCA) and RT-CES based NK cytotoxicity and analyzed degranulation activity (CD107a) and cytokine production. In 98 healthy individuals, FCA with peripheral blood mononuclear cells (PBMCs) at effector to target (E/T) ratio of 32 revealed 46.5 ± 2.6% cytolysis of K562 cells, and 23.5 ± 1.1% of NK cells showed increased degranulation. In RT-CES system, adherent NIH3T3 target cells were resistant to basal killing by PBMC or NK cells. NK cell activation by adding IL-2 demonstrated real-time dynamic killing activity, and lymphokine-activated PBMC (E/T ratio of 32) from 15 individuals showed 59.1 ± 6.2% cytotoxicity results after 4 hours incubation in RT-CES system. However, there was no significant correlation between FCA and RT-CES cytotoxicity. After K562 target cell stimulation, PBMC produced profound proinflammatory and immunoregulatory cytokines/chemokines including IL-2, IL-8, IL-10, MIP-1 α β , IFN- γ , and TNF- α , and cytokine/chemokine secretion was related to flowcytometry-based NK cytotoxicity. These data suggest that RT-CES and FCA differ in sensitivity, applicability and providing information, and further investigations are necessary in variable clinical conditions.
Collapse
Affiliation(s)
- Ki-Hyun Park
- Department of Biomedical Science, Graduate School, Catholic University of Korea, 505 Banpo-Dong, Seocho-Ku, Seoul 137-701, Republic of Korea
| | - Hyesun Park
- Department of Biomedical Science, Graduate School, Catholic University of Korea, 505 Banpo-Dong, Seocho-Ku, Seoul 137-701, Republic of Korea
| | - Myungshin Kim
- Department of Laboratory Medicine, Seoul St. Mary's Hospital, Catholic University of Korea, 505 Banpo-Dong, Seocho-Ku, Seoul 137-701, Republic of Korea
| | - Yonggoo Kim
- Department of Laboratory Medicine, Seoul St. Mary's Hospital, Catholic University of Korea, 505 Banpo-Dong, Seocho-Ku, Seoul 137-701, Republic of Korea
| | - Kyungja Han
- Department of Laboratory Medicine, Seoul St. Mary's Hospital, Catholic University of Korea, 505 Banpo-Dong, Seocho-Ku, Seoul 137-701, Republic of Korea
| | - Eun-Jee Oh
- Department of Laboratory Medicine, Seoul St. Mary's Hospital, Catholic University of Korea, 505 Banpo-Dong, Seocho-Ku, Seoul 137-701, Republic of Korea
| |
Collapse
|
35
|
Al-Ahmad A, Laird D, Zou P, Tomakidi P, Steinberg T, Lienkamp K. Nature-inspired antimicrobial polymers--assessment of their potential for biomedical applications. PLoS One 2013; 8:e73812. [PMID: 24040079 PMCID: PMC3767731 DOI: 10.1371/journal.pone.0073812] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 07/25/2013] [Indexed: 12/25/2022] Open
Abstract
We explored the potential of poly(oxonorbornene)-based synthetic mimics of antimicrobial peptides (SMAMPs), a promising new class of antimicrobial polymers with cell-selectivity and low resistance development potential, for clinical applications. We evaluated their antimicrobial activity against a panel of seven clinical and regulatory relevant bacteria strains, and tested their toxicity with two different kinds of primary human cells. For the antimicrobial activity, we performed the minimum inhibitory concentration (MIC) assay and determined the minimum bactericidal concentration (MBC) according to the NCCLS guidelines. The results revealed specific problems that may occur when testing the antimicrobial activity of amphiphilic cationic polymers, and confirmed the working hypothesis that the more hydrophilic SMAMP polymers in our portfolio were 'doubly selective', i.e. they are not only selective for bacteria over mammalian cells, but also for Gram-positive over Gram-negative bacteria. The data also showed that we could improve the broad-band activity of one SMAMP, and in combination with the results from the cell toxicity experiments, identified this polymer as a promising candidate for further in-vitro and in-vivo testing. Transmission electron studies revealed that the cellular envelopes of both E. coli and S. aureus were severely damaged due to SMAMP action on the bacterial membrane, which strengthened the argument that SMAMPs closely resemble antimicrobial peptides. To test cell toxicity, we used the traditional hemolysis assay with human red blood cells, and the novel xCelligence assay with primary human fibroblasts. The data reported here is the first example in which a hemolysis assay is benchmarked against the xCelligence assay. It revealed that the same trends were obtained using these complementary methods. This establishes the xCelligence assay with primary human cells as a useful tool for SMAMP characterization.
Collapse
Affiliation(s)
- Ali Al-Ahmad
- Operative Dentistry and Periodontology, University Medical Center of the Albert-Ludwigs-Universität, Freiburg, Germany
| | - Dougal Laird
- Freiburg Institute for Advanced Studies, Albert-Ludwigs-Universität, Freiburg, Germany
- Oral Biotechnology, University Medical Center of the Albert-Ludwigs-Universität, Freiburg, Germany
| | - Peng Zou
- Freiburg Institute for Advanced Studies, Albert-Ludwigs-Universität, Freiburg, Germany
- Department of Microsystems Engineering, Albert-Ludwigs-Universität, Freiburg, Germany
| | - Pascal Tomakidi
- Oral Biotechnology, University Medical Center of the Albert-Ludwigs-Universität, Freiburg, Germany
| | - Thorsten Steinberg
- Oral Biotechnology, University Medical Center of the Albert-Ludwigs-Universität, Freiburg, Germany
| | - Karen Lienkamp
- Freiburg Institute for Advanced Studies, Albert-Ludwigs-Universität, Freiburg, Germany
- Department of Microsystems Engineering, Albert-Ludwigs-Universität, Freiburg, Germany
| |
Collapse
|
36
|
M1 muscarinic receptor activation mediates cell death in M1-HEK293 cells. PLoS One 2013; 8:e72011. [PMID: 24023725 PMCID: PMC3759376 DOI: 10.1371/journal.pone.0072011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Accepted: 07/10/2013] [Indexed: 01/10/2023] Open
Abstract
HEK293 cells have been used extensively to generate stable cell lines to study G protein-coupled receptors, such as muscarinic acetylcholine receptors (mAChRs). The activation of M1 mAChRs in various cell types in vitro has been shown to be protective. To further investigate M1 mAChR-mediated cell survival, we generated stable HEK293 cell-lines expressing the human M1 mAChR. M1 mAChRs were efficiently expressed at the cell surface and efficiently internalised within 1 h by carbachol. Carbachol also induced early signalling cascades similar to previous reports. Thus, ectopically expressed M1 receptors behaved in a similar fashion to the native receptor over short time periods of analysis. However, substantial cell death was observed in HEK293-M1 cells within 24 h after carbachol application. Death was only observed in HEK cells expressing M1 receptors and fully blocked by M1 antagonists. M1 mAChR-stimulation mediated prolonged activation of the MEK-ERK pathway and resulted in prolonged induction of the transcription factor EGR-1 (>24 h). Blockade of ERK signalling with U0126 did not reduce M1 mAChR-mediated cell-death significantly but inhibited the acute induction of EGR-1. We investigated the time-course of cell death using time-lapse microscopy and xCELLigence technology. Both revealed the M1 mAChR cytotoxicity occurs within several hours of M1 activation. The xCELLigence assay also confirmed that the ERK pathway was not involved in cell-death. Interestingly, the MEK blocker did reduce carbachol-mediated cleaved caspase 3 expression in HEK293-M1 cells. The HEK293 cell line is a widely used pharmacological tool for studying G-protein coupled receptors, including mAChRs. Our results highlight the importance of investigating the longer term fate of these cells in short term signalling studies. Identifying how and why activation of the M1 mAChR signals apoptosis in these cells may lead to a better understanding of how mAChRs regulate cell-fate decisions.
Collapse
|
37
|
Teng Z, Kuang X, Wang J, Zhang X. Real-time cell analysis--a new method for dynamic, quantitative measurement of infectious viruses and antiserum neutralizing activity. J Virol Methods 2013; 193:364-70. [PMID: 23835032 DOI: 10.1016/j.jviromet.2013.06.034] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Revised: 06/19/2013] [Accepted: 06/26/2013] [Indexed: 11/19/2022]
Abstract
A newly developed electronic cell sensor array--the xCELLigence real-time cell analysis (RTCA) system is tested currently for dynamic monitoring of cell attachment, proliferation, damage, and death. In this study, human enterovirus (HEV71) infection of human rhabdomyosarcoma (RD) was used as an in vitro model to validate the application of this novel system as a straightforward and efficient assay for quantitative measurement of infectious viruses based on virus-induced cytopathic effect (CPE). Several experimental tests were performed including the determination of optimal seeding density of the RD cells in 96-well E-plates, RTCA real-time monitoring of the virus induced CPE and virus titer calculation, and viral neutralization test to determine HEV71 antibody titer. Traditional 50% tissue culture infective dose (TCID50) assay was also conducted for methodology comparison and validation, which indicated a consistent result between the two assays. These findings indicate that the xCELLigence RTCA system can be a valuable addition to current viral assays for quantitative measurement of infectious viruses and quantitation of neutralization antibody titer in real-time, warranting for future research and exploration of applications to many other animal and human viruses.
Collapse
Affiliation(s)
- Zheng Teng
- Department of Microbiology, Shanghai Municipal Center for Disease Control and Prevention, Shanghai 200336, China
| | | | | | | |
Collapse
|
38
|
Renner NA, Sansing HA, Inglis FM, Mehra S, Kaushal D, Lackner AA, Maclean AG. Transient acidification and subsequent proinflammatory cytokine stimulation of astrocytes induce distinct activation phenotypes. J Cell Physiol 2013; 228:1284-94. [PMID: 23154943 DOI: 10.1002/jcp.24283] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Accepted: 11/02/2012] [Indexed: 01/18/2023]
Abstract
The foot processes of astrocytes cover over 60% of the surface of brain microvascular endothelial cells, regulating tight junction integrity. Retraction of astrocyte foot processes has been postulated to be a key mechanism in pathology. Therefore, movement of an astrocyte in response to a proinflammatory cytokine or even limited retraction of processes would result in leaky junctions between endothelial cells. Astrocytes lie at the gateway to the CNS and are instrumental in controlling leukocyte entry. Cultured astrocytes typically have a polygonal morphology until stimulated. We hypothesized that cultured astrocytes which were induced to stellate would have an activated phenotype compared with polygonal cells. We investigated the activation of astrocytes derived from adult macaques to the cytokine TNF-α under resting and stellated conditions by four parameters: morphology, intermediate filament expression, adhesion, and cytokine secretion. Astrocytes were stellated following transient acidification; resulting in increased expression of GFAP and vimentin. Stellation was accompanied by decreased adhesion that could be recovered with proinflammatory cytokine treatment. Surprisingly, there was decreased secretion of proinflammatory cytokines by stellated astrocytes compared with polygonal cells. These results suggest that astrocytes are capable of multiple phenotypes depending on the stimulus and the order stimuli are applied.
Collapse
Affiliation(s)
- Nicole A Renner
- Program in Neuroscience, Tulane University, New Orleans, LA, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Cimpan MR, Mordal T, Schölermann J, Allouni ZE, Pliquett U, Cimpan E. An impedance-based high-throughput method for evaluating the cytotoxicity of nanoparticles. ACTA ACUST UNITED AC 2013. [DOI: 10.1088/1742-6596/429/1/012026] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
40
|
Evaluation of impedance-based label-free technology as a tool for pharmacology and toxicology investigations. BIOSENSORS-BASEL 2013; 3:132-56. [PMID: 25587404 PMCID: PMC4263586 DOI: 10.3390/bios3010132] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2013] [Revised: 02/15/2013] [Accepted: 02/27/2013] [Indexed: 12/23/2022]
Abstract
The use of label-free technologies based on electrical impedance is becoming more and more popular in drug discovery. Indeed, such a methodology allows the continuous monitoring of diverse cellular processes, including proliferation, migration, cytotoxicity and receptor-mediated signaling. The objective of the present study was to further assess the usefulness of the real-time cell analyzer (RTCA) and, in particular, the xCELLigence platform, in the context of early drug development for pharmacology and toxicology investigations. In the present manuscript, four cellular models were exposed to 50 compounds to compare the cell index generated by RTCA and cell viability measured with a traditional viability assay. The data revealed an acceptable correlation (ca. 80%) for both cell lines (i.e., HepG2 and HepaRG), but a lack of correlation (ca. 55%) for the primary human and rat hepatocytes. In addition, specific RTCA profiles (signatures) were generated when HepG2 and HepaRG cells were exposed to calcium modulators, antimitotics, DNA damaging and nuclear receptor agents, with a percentage of prediction close to 80% for both cellular models. In a subsequent experiment, HepG2 cells were exposed to 81 proprietary UCB compounds known to be genotoxic or not. Based on the DNA damaging signatures, the RTCA technology allowed the detection of ca. 50% of the genotoxic compounds (n = 29) and nearly 100% of the non-genotoxic compounds (n = 52). Overall, despite some limitations, the xCELLigence platform is a powerful and reliable tool that can be used in drug discovery for toxicity and pharmacology studies.
Collapse
|
41
|
Abstract
Demyelinating diseases such as multiple sclerosis are chronic inflammatory autoimmune diseases with a heterogeneous clinical presentation and course. Both the adaptive and the innate immune systems have been suggested to contribute to their pathogenesis and recovery. In this review, we discuss the role of the innate immune system in mediating demyelinating diseases. In particular, we provide an overview of the anti-inflammatory or pro-inflammatory functions of dendritic cells, mast cells, natural killer (NK) cells, NK-T cells, γδ T cells, microglial cells, and astrocytes. We emphasize the interaction of astroctyes with the immune system and how this interaction relates to the demyelinating pathologies. Given the pivotal role of the innate immune system, it is possible that targeting these cells may provide an effective therapeutic approach for demyelinating diseases.
Collapse
Affiliation(s)
- Lior Mayo
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | | |
Collapse
|
42
|
Ahmadi Z, Arababadi MK, Hassanshahi G. CXCL10 Activities, Biological Structure, and Source Along with Its Significant Role Played in Pathophysiology of Type I Diabetes Mellitus. Inflammation 2012; 36:364-71. [DOI: 10.1007/s10753-012-9555-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
43
|
Guan N, Gao W, He M, Zheng M, Xu X, Wang X, Wang MW. Dynamic monitoring of β-cell injury with impedance and rescue by glucagon-like peptide-1. Anal Biochem 2012; 423:61-9. [DOI: 10.1016/j.ab.2012.01.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Revised: 01/15/2012] [Accepted: 01/17/2012] [Indexed: 01/04/2023]
|
44
|
Burkert K, Moodley K, Angel CE, Brooks A, Graham ES. Detailed analysis of inflammatory and neuromodulatory cytokine secretion from human NT2 astrocytes using multiplex bead array. Neurochem Int 2011; 60:573-80. [PMID: 21939706 DOI: 10.1016/j.neuint.2011.09.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Revised: 08/12/2011] [Accepted: 09/05/2011] [Indexed: 11/26/2022]
Abstract
Astrocytes are a very important cell type in the brain fulfilling roles in both neuroimmunology and neurotransmission. We have conducted the most comprehensive analysis of secreted cytokines conducted to date (astrocytes of any source) to determine whether astrocytes derived from the human Ntera2 (NT2) cell-line are a good model of human primary astrocytes. We have compared the secretion of cytokines from NT2 astrocytes with those produced in astrocyte enriched human brain cultures and additional cytokines implicated in brain injury or known to be expressed in the human brain. The concentration of cytokines was measured in astrocyte conditioned media using multiplex bead array (MBA), where 18 cytokines were measured simultaneously. Resting NT2 astrocytes produced low levels (∼1-30 pg/ml) of MIP1α, IL-6 and GM-CSF and higher levels of MCP-1, IP-10 and IL-8 (1-11 ng/ml) under non-inflammatory conditions. All of these in addition to IL-1β, TNFα, and IL-13, were increased by pro-inflammatory activation (TNFα or IL-1β stimulation). In contrast, IL-2, IL-4, IL-5, IL-7, IL-10, IL-12, LTα, and IFNγ were not detected in astrocyte conditioned media under any of the culture conditions tested. NT2 astrocytes were unresponsive to IL-2 and the adenyl cyclase agonist, forskolin. Interestingly, IFNγ stimulation selectively increased IP-10 secretion only. As astrocytes stimulated with IL-1β or TNFα produced several chemokines in the ng/ml range, we next assessed the chemoattractant properties of these cells. Conditioned media from TNFα-stimulated astrocytes significantly chemoattracted leukocytes from human blood. This study provides the most comprehensive analysis of cytokine production by human astrocytes thus far, and shows that NT2 astrocytes are highly responsive to pro-inflammatory mediators including TNFα and IL-1β, producing cytokines and chemokines capable of attracting leukocytes from human blood. We conclude that in the absence of adult human primary astrocytes that NT2-astrocytes may provide a valuable alternative to study the immunological behaviour of human astrocytes.
Collapse
Affiliation(s)
- Kristina Burkert
- School of Biological Sciences, Faculty of Science, University of Auckland, New Zealand
| | | | | | | | | |
Collapse
|