1
|
Prša P, Miller IP, Kramar B, Šuput D, Milisav I. Short-Term Fasting Induces Hepatocytes' Stress Response and Increases Their Resilience. Int J Mol Sci 2025; 26:999. [PMID: 39940770 PMCID: PMC11817670 DOI: 10.3390/ijms26030999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 01/20/2025] [Accepted: 01/23/2025] [Indexed: 02/16/2025] Open
Abstract
Fasting leads to a range of metabolic adaptations that have developed through evolution, as humans and other mammals have unequal access to food over the circadian cycle and are therefore adapted to fasting and feeding cycles. We have investigated the role of a single fasting episode in rats in triggering the stress response of liver hepatocytes. Since the stress responses were observed in both animals and isolated cells, we investigated whether the effects of the animal stressor could persist in the cells after isolation. By measuring staurosporine-induced apoptosis, stress signalling, and oxidative and antioxidant responses in hepatocytes from fasted and ad libitum-fed animals, we found that only fasting animals elicited a stress response that prevented caspase-9 activation and persisted in isolated cells. The addition of glucose oxidase, a hydrogen peroxide-producing enzyme, to the cells from ad libitum-fed animals also led to a stress response phenotype and prevented the activation of caspase-9. A single fasting episode thus leads to a stress response in normal hepatocytes, with hydrogen peroxide as a second messenger that reduces the initiation of apoptosis. This finding is the first characterisation of a mechanism underlying the effects of fasting and provides a basis for the development of methods to increase the resilience of cells. These findings need to be taken into account when interpreting the results obtained in animal and cell research models to account for the effects of overnight fasting used in many laboratory protocols. The research results also form the basis for the development of clinical applications to increase the resistance of transplants and to improve the fitness of hepatocytes under acute stress conditions in liver and some metabolic diseases.
Collapse
Affiliation(s)
- Patrik Prša
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloska 4, SI-1000 Ljubljana, Slovenia
| | - Izak Patrik Miller
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloska 4, SI-1000 Ljubljana, Slovenia
| | - Barbara Kramar
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloska 4, SI-1000 Ljubljana, Slovenia
| | - Dušan Šuput
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloska 4, SI-1000 Ljubljana, Slovenia
| | - Irina Milisav
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloska 4, SI-1000 Ljubljana, Slovenia
- Laboratory of Oxidative Stress Research, Faculty of Health Sciences, University of Ljubljana, Zdravstvena pot 5, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
2
|
Gillespie KP, Pirnie R, Mesaros C, Blair IA. Cisplatin Dependent Secretion of Immunomodulatory High Mobility Group Box 1 (HMGB1) Protein from Lung Cancer Cells. Biomolecules 2023; 13:1335. [PMID: 37759736 PMCID: PMC10526420 DOI: 10.3390/biom13091335] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/25/2023] [Accepted: 08/26/2023] [Indexed: 09/29/2023] Open
Abstract
High mobility group box 1 (HMGB1) is secreted from activated immune cells, necrotic cells, and certain cancers. Previous studies have reported that different patterns of post-translational modification, particularly acetylation and oxidation, mediate HMGB1 release and confer distinct extracellular HMGB1 signaling activity. Here we report that cisplatin but not carboplatin induces secretion of HMGB1 from human A549 non-small cell lung cancer (NSCLC) cells. Cisplatin-mediated HMGB1 secretion was dose-dependent and was regulated by nuclear exportin 1 (XPO1) also known as chromosomal maintenance 1 (CRM1) rather than adenosine diphosphate (ADP)-ribosylation, acetylation, or oxidation. HMGB1, as well as lysine acetylation and cysteine disulfide oxidation of secreted HMGB1, were monitored by sensitive and specific assays using immunoprecipitation, stable isotope dilution, differential alkylation, and nano liquid chromatography parallel reaction monitoring/high-resolution mass spectrometry (nano-LC-PRM/HRMS). A major fraction of the HMGB1 secreted by low-dose cisplatin treatment of A549 NSCLC cells was found to be in the fully reduced form. In contrast, mainly oxidized forms of HMGB1 were secreted by dimethyl sulfoxide (DMSO)-mediated apoptosis. These findings suggest that inhibition of XPO1 could potentiate the anti-tumor activity of cisplatin by increasing the nuclear accumulation of HMGB1 protein, an inhibitor of cisplatin DNA-adduct repair. Furthermore, low-dose cisplatin therapy could modulate the immune response in NSCLC through the established chemokine activity of extracellular reduced HMGB1. This could potentially enhance the efficacy of subsequent immunotherapy treatment.
Collapse
Affiliation(s)
| | | | | | - Ian A. Blair
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
3
|
Kramar B, Šuput D, Milisav I. Differential p16 expression levels in the liver, hepatocytes and hepatocellular cell lines. PeerJ 2021; 9:e12358. [PMID: 34760375 PMCID: PMC8570159 DOI: 10.7717/peerj.12358] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 09/30/2021] [Indexed: 12/13/2022] Open
Abstract
Background One of the most frequently deleted genes in cancer is CDKN2A encoding p16. This protein is often overexpressed in senescent cells, while its suppression can bypass the oncogene-induced senescence to enable transformation and tumorigenesis. The roles of the protein p16 are recently being expanded from the cell cycle progression regulator to the cellular regulator interacting in several different pathways. Yet data on its liver and liver cells' expression are inconclusive. Methods The expression of the p16 gene in liver and liver cells was determined by RT-qPCR and compared to its protein amounts by western blotting. Results p16 is expressed at low levels in the liver and rat hepatocytes. Its expression varies from none to the considerable levels in the examined hepatocellular carcinoma cell lines (FaO and HepG2) and in immortalized mouse hepatocytes. Such significant expression differences of an important cellular regulator warrant the need to closely examine the differences in biochemical pathways correlated with the p16 expression when using hepatocytes and hepatoma liver models.
Collapse
Affiliation(s)
- Barbara Kramar
- University of Ljubljana, Faculty of Medicine, Institute of Pathophysiology, Zaloska 4, Ljubljana, Slovenia
| | - Dušan Šuput
- University of Ljubljana, Faculty of Medicine, Institute of Pathophysiology, Zaloska 4, Ljubljana, Slovenia
| | - Irina Milisav
- University of Ljubljana, Faculty of Medicine, Institute of Pathophysiology, Zaloska 4, Ljubljana, Slovenia.,University of Ljubljana, Laboratory of oxidative stress research, Faculty of Health Sciences, Zdravstvena pot 5, Ljubljana, Slovenia
| |
Collapse
|
4
|
Zhong Y, McGrath JK, Gong B. Dipropinonates of Sugar Alcohols as Water-Soluble, Nontoxic CPAs for DMSO-Free Cell Cryopreservation. ACS Biomater Sci Eng 2021; 7:4757-4762. [PMID: 34587440 DOI: 10.1021/acsbiomaterials.1c00995] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Sorbitol, mannitol, xylitol, and erythritol, four readily available sugar alcohols with poor or no membrane permeability, are converted into their corresponding dipropionates by acylating their primary hydroxyl groups. With enhanced membrane permeability, these diesters are expected to permeate the cell membranes and, upon their hydrolysis, release the corresponding sugar alcohols inside the cells. NIH-3T3 cells incubated with these diesters before being frozen at -80 °C exhibited considerably higher total recovery over those incubated with the free sugar alcohols or media only. Among the four diesters, those of sorbitol, especially mannitol, showed cryoprotective effects comparable to that shown by 5% DMSO. This work has demonstrated the feasibility of converting readily available, naturally occurring compounds into membrane-permeable derivatives that serve as water-soluble, nontoxic alternatives to DMSO.
Collapse
Affiliation(s)
- Yulong Zhong
- The State University of New York at Buffalo, Department of Chemistry, Buffalo, New York 14260, United States
| | - Jillian K McGrath
- The State University of New York at Buffalo, Department of Chemistry, Buffalo, New York 14260, United States
| | - Bing Gong
- The State University of New York at Buffalo, Department of Chemistry, Buffalo, New York 14260, United States
| |
Collapse
|
5
|
Wei F, Zhao L, Jing Y. Mechanisms underlying dimethyl sulfoxide-induced cellular migration in human normal hepatic cells. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2020; 80:103489. [PMID: 32911099 DOI: 10.1016/j.etap.2020.103489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 08/03/2020] [Accepted: 09/04/2020] [Indexed: 06/11/2023]
Abstract
Numerous studies have reported that low-dose dimethyl sulfoxide (DMSO, <1.5%, v/v) can interfere with various cellular processes, such as cell proliferation, differentiation, apoptosis, and cycle. By contrast, minimal information is available about the effect of low-dose DMSO on cell migration. Here, we report the effect of DMSO (0.0005%-0.5%, v/v) on cellular migration in human normal hepatic L02 cells. We used the Cell Counting Kit-8 assay to measure cell viability, scratch wound healing assay to observe cellular migration, flow cytometry to analyze cell cycle and death pattern, reverse transcription quantitative polymerase chain reaction to evaluate mRNA expression, and Western blot to detect protein levels. After treatment with 0.0005% (v/v) DMSO, more cells entered S phase arrest, the MMP1/TIMP1 ratio increased, and HSP27 expression was elevated. After treatment with 0.1% (v/v) DMSO, more cells entered G0/G1 phase arrest, the MMP2/TIMP2 ratio increased, the p-p38 and p-Smad3 signaling pathways were activated, and neuropilin-1 expression was elevated. These results showed that cells migrate when their MMP1/TIMP1 and MMP2/TIMP2 ratios are imbalanced. Such migration is modulated by the p38/HSP27 signaling pathway and TGF-β/Smad3 dependent signaling pathway.
Collapse
Affiliation(s)
- Fengmei Wei
- Department of Physiology and Psychology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu Province 730000, PR China
| | - Long Zhao
- Department of Orthopaedics, Lanzhou University First Affiliated Hospital, Lanzhou, Gansu Province 730000, PR China
| | - Yuhong Jing
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, 730000, PR China; Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou, Gansu, 730000, PR China.
| |
Collapse
|
6
|
Ayo-Lawal RA, Osoniyi O, Sibuyi NRS, Meyer M, Ekpo O. Cytotoxic and Apoptotic Induction Potential of Extracts from Fermented Citrullus vulgaris Thunb. Seeds on Cervical and Liver Cancer Cells. J Diet Suppl 2020; 18:132-146. [PMID: 32114858 DOI: 10.1080/19390211.2020.1731045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
The anti-cancer activities of many fermented foods and beverages are now scientifically established. Ogiri-egusi is a condiment prepared from fermentation of Citrullus vulgaris (melon) seeds and consumed in many countries of West Africa. Its anti-oxidative and anti-hyperlipidaemic properties have been reported. This study investigated the anti-cancer activities of the aqueous and methanolic extracts from ogiri-egusi. Cytotoxicity was investigated using the MTT and colony-formation inhibition assays while flow-cytometer based Apopercentage assay was used to quantify apoptosis in extracts-treated cervical and liver cancer and normal human fibroblast cells. The inhibitory concentration responsible for killing 50% of cells after 24 h by the aqueous extract in KMST-6, HeLa, and Hep-G2 cells were estimated at 1.610, 1.020, and 1.507 mg/mL respectively. While these values reduced with increasing incubation time in cancer cells it increased in the non-cancer cell. Furthermore, the extract significantly induced apoptosis in HeLa (97 ± 0.18%) and Hep-G2 (73 ± 6.73%) cells. These findings were corroborated by cells morphologic presentations and inhibition of colony formation assay. These findings suggest that the aqueous extract from fermented Citrullus vulgaris seeds might be a nutraceutical with potential anti-cancer properties.
Collapse
Affiliation(s)
- Rachael Aderonke Ayo-Lawal
- National Centre for Technology Management (NACETEM), Obafemi Awolowo University, Ile-Ife, Nigeria.,Department of Biochemistry and Molecular Biology, Obafemi Awolowo University, Ile-Ife, Nigeria
| | - Omolaja Osoniyi
- Department of Biochemistry and Molecular Biology, Obafemi Awolowo University, Ile-Ife, Nigeria
| | - Nicole Remaliah Samantha Sibuyi
- Department of Science and Technology/Mintek Nanotechnology Innovation Centre, Biolabels Unit, Department of Biotechnology, University of the Western Cape, Bellville, Cape Town, South Africa
| | - Mervin Meyer
- Department of Science and Technology/Mintek Nanotechnology Innovation Centre, Biolabels Unit, Department of Biotechnology, University of the Western Cape, Bellville, Cape Town, South Africa
| | - Okobi Ekpo
- Department of Medical Bioscience, University of the Western Cape, Bellville, Cape Town, South Africa
| |
Collapse
|
7
|
Awan M, Buriak I, Fleck R, Fuller B, Goltsev A, Kerby J, Lowdell M, Mericka P, Petrenko A, Petrenko Y, Rogulska O, Stolzing A, Stacey GN. Dimethyl sulfoxide: a central player since the dawn of cryobiology, is efficacy balanced by toxicity? Regen Med 2020; 15:1463-1491. [PMID: 32342730 DOI: 10.2217/rme-2019-0145] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Dimethyl sulfoxide (DMSO) is the cryoprotectant of choice for most animal cell systems since the early history of cryopreservation. It has been used for decades in many thousands of cell transplants. These treatments would not have taken place without suitable sources of DMSO that enabled stable and safe storage of bone marrow and blood cells until needed for transfusion. Nevertheless, its effects on cell biology and apparent toxicity in patients have been an ongoing topic of debate, driving the search for less cytotoxic cryoprotectants. This review seeks to place the toxicity of DMSO in context of its effectiveness. It will also consider means of reducing its toxic effects, the alternatives to its use and their readiness for active use in clinical settings.
Collapse
Affiliation(s)
- Maooz Awan
- Institute for Liver & Digestive Health, UCL Division of Medicine, Royal Free Hospital, UCL, London, NW3 2PF, UK
| | - Iryna Buriak
- Institute for Problems of Cryobiology & Cryomedicine, National Academy of Sciences of Ukraine, Pereyaslavska 23, 61016, Kharkiv
| | - Roland Fleck
- Centre for Ultrastructural Imaging, Kings College London, London, SE1 1UL, UK
| | - Barry Fuller
- Department of Surgical Biotechnology, UCL Division of Surgery, Royal Free Hospital, UCL, London, NW3 2QG, UK
| | - Anatoliy Goltsev
- Institute for Problems of Cryobiology & Cryomedicine, National Academy of Sciences of Ukraine, Pereyaslavska 23, 61016, Kharkiv
| | - Julie Kerby
- Cell & Gene Therapy Catapult, 12th Floor Tower Wing, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Mark Lowdell
- Centre for Cell, Gene & Tissue Therapy, Royal Free London NHS FT & UCL, London, NW3 2PF, UK
| | - Pavel Mericka
- Tissue Bank, University Hospital Hradec Kralové, Czech Republic
| | - Alexander Petrenko
- Institute for Problems of Cryobiology & Cryomedicine, National Academy of Sciences of Ukraine, Pereyaslavska 23, 61016, Kharkiv
| | - Yuri Petrenko
- Department of Biomaterials & Biophysical Methods, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
| | - Olena Rogulska
- Institute for Problems of Cryobiology & Cryomedicine, National Academy of Sciences of Ukraine, Pereyaslavska 23, 61016, Kharkiv
| | - Alexandra Stolzing
- University of Loughborough, Centre for Biological Engineering, Loughborough University, Holywell Park, Loughborough, UK
| | - Glyn N Stacey
- International Stem Cell Banking Initiative, 2 High Street, Barley, Hertfordshire, SG8 8HZ
- Beijing Stem Cell Bank, Institute of Zoology, Chinese Academy of Sciences, 25–2 Beishuan West, Haidan District, 100190 Beijing, China
- Institute of Stem Cells & Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
8
|
Pekkala S, Keskitalo A, Kettunen E, Lensu S, Nykänen N, Kuopio T, Ritvos O, Hentilä J, Nissinen TA, Hulmi JJ. Blocking Activin Receptor Ligands Is Not Sufficient to Rescue Cancer-Associated Gut Microbiota-A Role for Gut Microbial Flagellin in Colorectal Cancer and Cachexia? Cancers (Basel) 2019; 11:cancers11111799. [PMID: 31731747 PMCID: PMC6896205 DOI: 10.3390/cancers11111799] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 11/08/2019] [Accepted: 11/10/2019] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) and cachexia are associated with the gut microbiota and microbial surface molecules. We characterized the CRC-associated microbiota and investigated whether cachexia affects the microbiota composition. Further, we examined the possible relationship between the microbial surface molecule flagellin and CRC. CRC cells (C26) were inoculated into mice. Activin receptor (ACVR) ligands were blocked, either before tumor formation or before and after, to increase muscle mass and prevent muscle loss. The effects of flagellin on C26-cells were studied in vitro. The occurrence of similar phenomena were studied in murine and human tumors. Cancer modulated the gut microbiota without consistent effects of blocking the ACVR ligands. However, continued treatment for muscle loss modified the association between microbiota and weight loss. Several abundant microbial taxa in cancer were flagellated. Exposure of C26-cells to flagellin increased IL6 and CCL2/MCP-1 mRNA and IL6 excretion. Murine C26 tumors expressed more IL6 and CCL2/MCP-1 mRNA than C26-cells, and human CRC tumors expressed more CCL2/MCP-1 than healthy colon sites. Additionally, flagellin decreased caspase-1 activity and the production of reactive oxygen species, and increased cytotoxicity in C26-cells. Conditioned media from flagellin-treated C26-cells deteriorated C2C12-myotubes and decreased their number. In conclusion, cancer increased flagellated microbes that may promote CRC survival and cachexia by inducing inflammatory proteins such as MCP-1. Cancer-associated gut microbiota could not be rescued by blocking ACVR ligands.
Collapse
Affiliation(s)
- Satu Pekkala
- Faculty of Sport and Health Sciences, University of Jyväskylä, 40620 Jyväskylä, Finland; (E.K.); (S.L.); (J.H.); (T.A.N.); (J.J.H.)
- Correspondence: ; Tel.: +358-45-358-2898
| | - Anniina Keskitalo
- Institute of Biomedicine, Faculty of Medicine, University of Turku, 20500 Turku, Finland;
- Department of Clinical Microbiology, Turku University Hospital, 20500 Turku, Finland
| | - Emilia Kettunen
- Faculty of Sport and Health Sciences, University of Jyväskylä, 40620 Jyväskylä, Finland; (E.K.); (S.L.); (J.H.); (T.A.N.); (J.J.H.)
| | - Sanna Lensu
- Faculty of Sport and Health Sciences, University of Jyväskylä, 40620 Jyväskylä, Finland; (E.K.); (S.L.); (J.H.); (T.A.N.); (J.J.H.)
| | - Noora Nykänen
- Department of Pathology, Central Finland Health Care District, Keskussairaalantie 19, 40620 Jyväskylä, Finland; (N.N.); (T.K.)
| | - Teijo Kuopio
- Department of Pathology, Central Finland Health Care District, Keskussairaalantie 19, 40620 Jyväskylä, Finland; (N.N.); (T.K.)
- Department of Biological and Environmental Science, University of Jyväskylä, 40620 Jyväskylä, Finland
| | - Olli Ritvos
- Department of Physiology, Faculty of Medicine, University of Helsinki, 00100 Helsinki, Finland;
| | - Jaakko Hentilä
- Faculty of Sport and Health Sciences, University of Jyväskylä, 40620 Jyväskylä, Finland; (E.K.); (S.L.); (J.H.); (T.A.N.); (J.J.H.)
| | - Tuuli A. Nissinen
- Faculty of Sport and Health Sciences, University of Jyväskylä, 40620 Jyväskylä, Finland; (E.K.); (S.L.); (J.H.); (T.A.N.); (J.J.H.)
| | - Juha J. Hulmi
- Faculty of Sport and Health Sciences, University of Jyväskylä, 40620 Jyväskylä, Finland; (E.K.); (S.L.); (J.H.); (T.A.N.); (J.J.H.)
| |
Collapse
|
9
|
Beneficial Role of ROS in Cell Survival: Moderate Increases in H 2O 2 Production Induced by Hepatocyte Isolation Mediate Stress Adaptation and Enhanced Survival. Antioxidants (Basel) 2019; 8:antiox8100434. [PMID: 31581418 PMCID: PMC6826461 DOI: 10.3390/antiox8100434] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 09/07/2019] [Accepted: 09/17/2019] [Indexed: 01/06/2023] Open
Abstract
High levels of reactive oxygen species (ROS) can lead to impairment of cell structure, biomolecules' loss of function and cell death and are associated with liver diseases. Cells that survive increased ROS often undergo malignant transformation. Many cancer cells tolerate high levels of ROS. Here we report a transiently increased production of H2O2 and concomitant upregulation of antioxidative enzymes triggered by hepatocyte isolation; the H2O2 levels revert in about two days in culture. Three-day survival rate of the isolated cells in the presence of 2.5-fold increase of H2O2 is almost 80%. Apoptosis activation through the mitochondrial pathway is meanwhile reduced by inhibition of caspase-9 triggering. This reduction depends on the amount of H2O2 production, as decreased production of H2O2 in the presence of an antioxidant results in increased apoptosis triggering. These stress adaptations do not influence urea production, which is unchanged throughout the normal and stress adapted phases. We conclude that hepatocytes' stress adaptation is mediated by increased ROS production. In this case, high ROS improve cell survival.
Collapse
|
10
|
Vilas-Boas V, Cooreman A, Gijbels E, Van Campenhout R, Gustafson E, Ballet S, Annaert P, Cogliati B, Vinken M. Primary hepatocytes and their cultures for the testing of drug-induced liver injury. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2018; 85:1-30. [PMID: 31307583 DOI: 10.1016/bs.apha.2018.08.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Drug-induced liver injury is a major reason for discontinuation of drug development and withdrawal of drugs from the market. Intensive efforts in the last decades have focused on the establishment and finetuning of liver-based in vitro models for reliable prediction of hepatotoxicity triggered by drug candidates. Of those, primary hepatocytes and their cultures still are considered the gold standard, as they provide an acceptable reflection of the hepatic in vivo situation. Nevertheless, these in vitro systems cope with gradual deterioration of the differentiated morphological and functional phenotype. The present paper gives an overview of traditional and more recently introduced strategies to counteract this dedifferentiation process in an attempt to set up culture models that can be used for long-term testing purposes. The relevance and applicability of such optimized cultures of primary hepatocytes for the testing of drug-induced cholestatic liver injury is demonstrated.
Collapse
Affiliation(s)
- Vânia Vilas-Boas
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Axelle Cooreman
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Eva Gijbels
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Raf Van Campenhout
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Emma Gustafson
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Steven Ballet
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Pieter Annaert
- Drug Delivery and Disposition, KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| | - Bruno Cogliati
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Mathieu Vinken
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
11
|
Bell R, Robles-Harris M, Anderson M, Laudier D, Schaffler M, Flatow E, Andarawis-Puri N. Inhibition of apoptosis exacerbates fatigue-damage tendon injuries in an in vivo rat model. Eur Cell Mater 2018; 36:44-56. [PMID: 30058060 PMCID: PMC6350530 DOI: 10.22203/ecm.v036a04] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Tendinopathy is a common and progressive musculoskeletal disease. Increased apoptosis is an end-stage tendinopathy manifestation, but its contribution to the pathology of the disease is unknown. A previously established in vivo model of fatigue damage accumulation shows that increased apoptosis is correlated with the severity of induced tendon damage, even in early onset of the disease, supporting its implication in the pathogenesis of the disease. Consequently, this study aimed to determine: (1) whether apoptosis could be inhibited after fatigue damage and (2) whether its inhibition could lead to remodeling of the extracellular matrix (ECM) and pericellular matrix (PCM), to ultimately improve the mechanical properties of fatigue-damaged tendons. The working hypothesis was that, despite the low vascular nature of the tendon, apoptosis would be inhibited, prompting increased production of matrix proteins and restoring tendon mechanical properties. Rats received 2 or 5 d of systemic pan-caspase inhibitor (Q-VD-OPh) or dimethyl sulfoxide (DMSO) carrier control injections starting immediately prior to fatigue loading and were sacrificed at days 7 and 14 post-fatigue-loading. Systemic pan-caspase inhibition for 2 d led to a surprising increase in apoptosis, but inhibition for 5 d increased the population of live cells that could repair the fatigue damage. Further analysis of the 5 d group showed that effective inhibition led to an increased population of cells producing ECM and PCM proteins, although typically in conjunction with oxidative stress markers. Ultimately, inhibition of apoptosis led to further deterioration in mechanical properties of fatigue-damaged tendons.
Collapse
Affiliation(s)
- R. Bell
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
| | - M.A. Robles-Harris
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - M. Anderson
- Leni and Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - D. Laudier
- Leni and Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - M.B. Schaffler
- Department of Biomedical Engineering, City College of New York, New York, NY, USA
| | - E.L. Flatow
- Leni and Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - N. Andarawis-Puri
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA,Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA,Hospital for Special Surgery, New York, NY, USA,Address for correspondence: Nelly Andarawis-Puri, PhD, Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, 14850, NY, USA.
| |
Collapse
|
12
|
Huang Y, Cartlidge R, Walpitagama M, Kaslin J, Campana O, Wlodkowic D. Unsuitable use of DMSO for assessing behavioral endpoints in aquatic model species. THE SCIENCE OF THE TOTAL ENVIRONMENT 2018; 615:107-114. [PMID: 28963892 DOI: 10.1016/j.scitotenv.2017.09.260] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 09/23/2017] [Accepted: 09/24/2017] [Indexed: 06/07/2023]
Abstract
Dimethyl sulfoxide (DMSO) is a universally used aprotic solvent with the ability to permeate biological membranes and thus is commonly used to achieve appropriate biological availability of hydrophobic toxicants. While DMSO as a carrier medium has a reportedly low toxicity and is routinely employed in ecotoxicology, very little is known about its effect on dynamic behavioral parameters. This study presents a comparative analysis of the lethal and behavioral effects of exposures to DMSO concentrations of 0.1-10% on several test species such as: neonates of the freshwater crustacean Daphnia magna, nauplii of the marine crustacean Artemia franciscana, the marine crustacean Allorchestes compressa, embryos and larvae of the freshwater fish Danio rerio. The results demonstrated that DMSO did not cause statistically significant mortality even at concentrations close to 1% but induced clear and significant behavioral abnormalities in response to sublethal concentrations on all test species. These included hypoactivity syndrome in A. franciscana, A. compressa, D. magna and zebrafish larvae while a slight time-dependent hyperactivity response was observed in zebrafish embryos. For the majority of test species, behavioral changes such as moving distance, acceleration and burst movement were often observed during the first hours of exposure. These results indicate that caution should be exercised when using DMSO as a carrier solvent in experiments assessing behavioral endpoints.
Collapse
Affiliation(s)
- Yushi Huang
- School of Science, RMIT University, Melbourne, VIC, Australia
| | - Rhys Cartlidge
- School of Science, RMIT University, Melbourne, VIC, Australia
| | | | - Jan Kaslin
- ARMI, Monash University, Wellington Rd, Clayton, VIC, Australia
| | - Olivia Campana
- Instituto de Ciencias Marinas de Andalucia, CSIC, Cadiz, Spain
| | - Donald Wlodkowic
- School of Science, RMIT University, Melbourne, VIC, Australia; Centre for Environmental Sustainability and Remediation (EnSuRe), RMIT University, Melbourne, VIC, Australia.
| |
Collapse
|
13
|
Milisav I, Banič B, Šuput D. Animal nutrition and breeding conditions modify the physiology of isolated primary cells. Med Hypotheses 2017; 102:16-18. [PMID: 28478822 DOI: 10.1016/j.mehy.2017.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 01/20/2017] [Accepted: 02/26/2017] [Indexed: 10/20/2022]
Abstract
Animal primary cell cultures are widely used in biomedical research to investigate cell metabolism, diseases and to devise novel treatments. Modern animal breeding techniques are developed to unify, control and reduce the amount of microorganisms that the animals are being exposed to. Furthermore, health monitoring and strict caging and handling protocols allow animals to be exposed only to a selected spectrum of microbes. We are starting to appreciate that nutrition can influence composition of gut microbiota that can impact hosting organism's physiology and can even result in development of pathological changes. Evidence is also emerging that acute as well as chronic stresses can profoundly influence the physiology of certain organs, especially heart and liver. Our preliminary data imply that changes in animal nutrition and stress levels initiated up to minutes before the cell isolation could alter the cell stress response of cultured primary hepatocytes after isolation, leading to differences in sensitivity of apoptosis triggering. Therefore, we propose the hypothesis that conditions of animal breeding, especially diet and stress levels, are reflected in the physiology of the isolated primary cells. Variations in animal breeding conditions may influence experimental results on isolated cells and their applicability for studying human disorders.
Collapse
Affiliation(s)
- Irina Milisav
- University of Ljubljana, Faculty of Medicine, Institute of Pathophysiology, Zaloška 4, SI-1000 Ljubljana, Slovenia; University of Ljubljana, Faculty of Health Sciences, Ljubljana, Slovenia.
| | - Blaž Banič
- University of Ljubljana, Faculty of Medicine, Institute of Pathophysiology, Zaloška 4, SI-1000 Ljubljana, Slovenia
| | - Dušan Šuput
- University of Ljubljana, Faculty of Medicine, Institute of Pathophysiology, Zaloška 4, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
14
|
Hyun SW, Kim BR, Hyun SA, Seo JW. The assessment of electrophysiological activity in human-induced pluripotent stem cell-derived cardiomyocytes exposed to dimethyl sulfoxide and ethanol by manual patch clamp and multi-electrode array system. J Pharmacol Toxicol Methods 2017; 87:93-98. [PMID: 28377112 DOI: 10.1016/j.vascn.2017.03.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 03/14/2017] [Accepted: 03/31/2017] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Recently, electrophysiological activity has been effectively measured in human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) to predict drug-induced arrhythmia. Dimethyl sulfoxide (DMSO) and ethanol have been used as diluting agents in many experiments. However, the maximum DMSO and ethanol concentrations that can be effectively used in the measurement of electrophysiological parameters in hiPSC-CMs-based patch clamp and multi-electrode array (MEA) have not been fully elucidated. METHODS We investigated the effects of varying concentrations of DMSO and ethanol used as diluting agents on several electrophysiological parameters in hiPSC-CMs using patch clamp and MEA. RESULTS Both DMSO and ethanol at concentrations>1% in external solution resulted in osmolality >400mOsmol/kg, but pH was not affected by either agent. Neither DMSO nor ethanol led to cell death at the concentrations examined. However, resting membrane potential, action potential amplitude, action potential duration at 90% and 40%, and corrected field potential duration were decreased significantly at 1% ethanol concentration. DMSO at 1% also significantly decreased the sodium spike amplitude. In addition, the waveform of action potential and field potential was recorded as irregular at 3% concentrations of both DMSO and ethanol. Concentrations of up to 0.3% of either agent did not affect osmolality, pH, cell death, or electrophysiological parameters in hiPSC-CMs. DISCUSSION Our findings suggest that 0.3% is the maximum concentration at which DMSO or ethanol should be used for dilution purposes in hiPSC-CMs-based patch clamp and MEA.
Collapse
Affiliation(s)
- Soo-Wang Hyun
- Research Group for Safety Pharmacology, Korea Institute of Toxicology, KRICT, 34114 Daejeon, Republic of Korea.
| | - Bo-Ram Kim
- Research Group for Safety Pharmacology, Korea Institute of Toxicology, KRICT, 34114 Daejeon, Republic of Korea.
| | - Sung-Ae Hyun
- Research Group for Safety Pharmacology, Korea Institute of Toxicology, KRICT, 34114 Daejeon, Republic of Korea.
| | - Joung-Wook Seo
- Research Group for Safety Pharmacology, Korea Institute of Toxicology, KRICT, 34114 Daejeon, Republic of Korea.
| |
Collapse
|
15
|
|
16
|
Furness S, Hare DL, Kourakis A, Turnley AM, Wookey PJ. A novel ligand of calcitonin receptor reveals a potential new sensor that modulates programmed cell death. Cell Death Discov 2016; 2:16062. [PMID: 27777788 PMCID: PMC5056446 DOI: 10.1038/cddiscovery.2016.62] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 06/14/2016] [Accepted: 07/14/2016] [Indexed: 11/24/2022] Open
Abstract
We have discovered that the accumulation of an anti-calcitonin receptor (anti-CTR) antibody conjugated to a fluorophore (mAb2C4:AF568) provides a robust signal for cells undergoing apoptotic programmed cell death (PCD). PCD is an absolute requirement for normal development of metazoan organisms. PCD is a hallmark of common diseases such as cardiovascular disease and tissue rejection in graft versus host pathologies, and chemotherapeutics work by increasing PCD. This robust signal or high fluorescent events were verified by confocal microscopy and flow cytometry in several cell lines and a primary culture in which PCD had been induced. In Jurkat cells, GBM-L2 and MG63 cells, the percentage undergoing PCD that were positive for both mAb2C4:AF568 and annexin V ranged between 70 and >90%. In MG63 cells induced for the preapoptotic cell stress response (PACSR), the normal expression of α-tubulin, a key structural component of the cytoskeleton, and accumulation of mAb2C4:AF568 were mutually exclusive. Our data support a model in which CTR is upregulated during PACSR and recycles to the plasma membrane with apoptosis. In cells committed to apoptosis (α-tubulin negative), there is accumulation of the CTR-ligand mAb2C4:AF568 generating a high fluorescent event. The reagent mAb2C4:AF568 effectively identifies a novel event linked to apoptosis.
Collapse
Affiliation(s)
- Sgb Furness
- Drug Discovery Biology Laboratory, Monash Institute of Pharmaceutical Science, 381 Royal Parade, Parkville, Victoria 3052, Australia; Department of Pharmacology, Monash University, Wellington Road, Clayton, Parkville, Victoria 3800, Australia
| | - D L Hare
- Department of Medicine, University of Melbourne, Austin Health , Level 7, Lance Townsend Building, Studley Road, Heidelberg, Victoria 3084, Australia
| | - A Kourakis
- Department of Medicine, University of Melbourne, Austin Health , Level 7, Lance Townsend Building, Studley Road, Heidelberg, Victoria 3084, Australia
| | - A M Turnley
- Department of Anatomy and Neuroscience, Melbourne Brain Centre, University of Melbourne , Royal Parade, Parkville, Victoria 3010, Australia
| | - P J Wookey
- Department of Medicine, University of Melbourne, Austin Health , Level 7, Lance Townsend Building, Studley Road, Heidelberg, Victoria 3084, Australia
| |
Collapse
|
17
|
Koller A, Rid R, Beyreis M, Bianchini R, Holub BS, Lang A, Sternberg F, Brodowicz B, Velickovic O, Jakab M, Kerschbaum H, Önder K, Kofler B. In vitro toxicity of the galanin receptor 3 antagonist SNAP 37889. Neuropeptides 2016; 56:83-8. [PMID: 26725588 DOI: 10.1016/j.npep.2015.12.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 12/04/2015] [Accepted: 12/06/2015] [Indexed: 12/20/2022]
Abstract
Galanin and its receptors (GAL1, GAL2, GAL3) modulate a range of neuronal, immune and vascular activities. In vivo administration of SNAP 37889 (1-phenyl-3-[[3-(trifluoromethyl)phenyl]imino]-1H-indol-2-one), a potent small non-peptidergic antagonist of GAL3, was reported to reduce anxiety- and depression-related behavior, ethanol consumption, and antagonizes the effect of galanin on plasma extravasation in rodent models. Accordingly, SNAP 37889 has been proposed as a potential therapeutic agent to treat anxiety and depression disorders. Therefore, we evaluated the toxicity of SNAP 37889 to different cell types. Our experiments revealed that SNAP 37889 (≥10μM) induced apoptosis in epithelial (HMCB) and microglial (BV-2) cell lines expressing endogenous GAL3, in peripheral blood mononuclear cells and promyelocytic leukemia cells (HL-60) expressing GAL2, and in a neuronal cell line (SH-SY5Y) lacking galanin receptor expression altogether. In conclusion, SNAP 37889 is toxic to a variety of cell types independent of GAL3 expression. We caution that the clinical use of SNAP 37889 at doses that might be used to treat anxiety- or depression- related diseases could have unexpected non-galanin receptor-mediated toxicity, especially on immune cells.
Collapse
Affiliation(s)
- Andreas Koller
- Laura Bassi Centre of Expertise THERAPEP, Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, Paracelsus Medical University, Müllner Hauptstraße 48, 5020 Salzburg, Austria
| | - Raphaela Rid
- Division of Molecular Dermatology, Department of Dermatology, Paracelsus Medical University, Müllner Hauptstraße 48, 5020 Salzburg, Austria
| | - Marlena Beyreis
- Department of Cell Biology, University of Salzburg, Hellbrunnerstraße 34, 5020 Salzburg, Austria; Institute of Physiology and Pathophysiology, Paracelsus Medical University, Strubergasse 21, 5020 Salzburg, Austria; Gastein Research Institute, Paracelsus Medical University, Strubergasse 21, 5020 Salzburg, Austria
| | - Rodolfo Bianchini
- Laura Bassi Centre of Expertise THERAPEP, Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, Paracelsus Medical University, Müllner Hauptstraße 48, 5020 Salzburg, Austria
| | - Barbara S Holub
- Laura Bassi Centre of Expertise THERAPEP, Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, Paracelsus Medical University, Müllner Hauptstraße 48, 5020 Salzburg, Austria
| | - Andreas Lang
- Laura Bassi Centre of Expertise THERAPEP, Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, Paracelsus Medical University, Müllner Hauptstraße 48, 5020 Salzburg, Austria
| | - Felix Sternberg
- Laura Bassi Centre of Expertise THERAPEP, Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, Paracelsus Medical University, Müllner Hauptstraße 48, 5020 Salzburg, Austria
| | - Bernhard Brodowicz
- Laura Bassi Centre of Expertise THERAPEP, Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, Paracelsus Medical University, Müllner Hauptstraße 48, 5020 Salzburg, Austria
| | - Ognjen Velickovic
- Division of Molecular Dermatology, Department of Dermatology, Paracelsus Medical University, Müllner Hauptstraße 48, 5020 Salzburg, Austria
| | - Martin Jakab
- Institute of Physiology and Pathophysiology, Paracelsus Medical University, Strubergasse 21, 5020 Salzburg, Austria
| | - Hubert Kerschbaum
- Department of Cell Biology, University of Salzburg, Hellbrunnerstraße 34, 5020 Salzburg, Austria
| | - Kamil Önder
- Division of Molecular Dermatology, Department of Dermatology, Paracelsus Medical University, Müllner Hauptstraße 48, 5020 Salzburg, Austria; Procomcure Biotech GmbH, Austria
| | - Barbara Kofler
- Laura Bassi Centre of Expertise THERAPEP, Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, Paracelsus Medical University, Müllner Hauptstraße 48, 5020 Salzburg, Austria.
| |
Collapse
|
18
|
Mitręga KA, Nożyński J, Porc M, Spałek AM, Krzemiński TF. Dihydropyridines' metabolites-induced early apoptosis after myocardial infarction in rats; new outlook on preclinical study with M-2 and M-3. Apoptosis 2016; 21:195-208. [PMID: 26666749 PMCID: PMC4712235 DOI: 10.1007/s10495-015-1205-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Our previous studies established cardio-protective effects of furnidipine and its active metabolites called M-2 and M-3. The aim of current research was to compare the effects of single oral pretreatment with 20 mg kg(-1) of M-2 and M-3 on mortality, different forms of arrhythmias, blood pressures parameters and ST-segment changes during occlusion (for 90 min) and reperfusion in the model of myocardial infarction in rats evoked by left anterior descending coronary artery occlusion. Additionally, the development of programmed cell death and biochemical parameters in blood serum were studied at 4th day after infarction. Furnidipines' metabolites effectively reduced mortality index while did not markedly influence on blood pressures parameters, arrhythmias, ST-segment changes as well as biochemical parameters. Intriguingly, programmed cell death study (TUNEL) showed distinct increase in the amount of apoptotic nuclei in post-infarcted myocardium, granulation tissue and what is more in arteriolar walls after M-2 and M-3 application. Moreover, M-2 turned out to be more powerful in stimulation of apoptosis in granulation tissue surrounding infarcted area whereas M-3 presented balanced profile in this matter. Taking into account that programmed cell death plays positive role in post-infarcted heart healing, M-2 presents itself as more attractive agent for oral pretreatment in early stages of ischemia by non-stable individuals due to its more specific action in stimulation repairing processes in granulation tissue as well as in arteriolar walls. While M-2 and M-3 are common metabolites present in degradation pathways of many widely used dihydropyridines in clinic, this key fact put the new outlook on understanding additional mechanism and effects of not only furnidipines' metabolites but also other dihydropyridines.
Collapse
Affiliation(s)
- Katarzyna A Mitręga
- Chair and Department of Pharmacology, Medical University of Silesia, ul. Jordana 19, 41-808, Zabrze, Poland
| | - Jerzy Nożyński
- Silesian Centre for Heart Diseases, ul. Szpitalna 2, 41-800, Zabrze, Poland
| | - Maurycy Porc
- Chair and Department of Pharmacology, Medical University of Silesia, ul. Jordana 19, 41-808, Zabrze, Poland
| | - Adrianna M Spałek
- Chair and Department of Pharmacology, Medical University of Silesia, ul. Jordana 19, 41-808, Zabrze, Poland.
| | - Tadeusz F Krzemiński
- Chair and Department of Pharmacology, Medical University of Silesia, ul. Jordana 19, 41-808, Zabrze, Poland
| |
Collapse
|
19
|
Abstract
High levels of penetrating cryoprotectants (CPAs) can eliminate ice formation during cryopreservation of cells, tissues, and organs to cryogenic temperatures. But CPAs become increasingly toxic as concentration increases. Many strategies have been attempted to overcome the problem of eliminating ice while minimizing toxicity, such as attempting to optimize cooling and warming rates, or attempting to optimize time of adding individual CPAs during cooling. Because strategies currently used are not adequate, CPA toxicity remains the greatest obstacle to cryopreservation. CPA toxicity stands in the way of cryogenic cryopreservation of human organs, a procedure that has the potential to save many lives. This review attempts to describe what is known about CPA toxicity, theories of CPA toxicity, and strategies to reduce CPA toxicity. Critical analysis and suggestions are also included.
Collapse
|
20
|
Solanas E, Sostres C, Serrablo A, García-Gil A, García JJ, Aranguren FJ, Jiménez P, Hughes RD, Serrano MT. Effect of Dimethyl Sulfoxide and Melatonin on the Isolation of Human Primary Hepatocytes. Cells Tissues Organs 2015; 200:316-25. [DOI: 10.1159/000433521] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/26/2015] [Indexed: 11/19/2022] Open
|
21
|
Milisav I, Ribarič S, Šuput D. Targeting stress responses for regenerative medicine. Methods Mol Biol 2015; 1292:235-43. [PMID: 25804760 DOI: 10.1007/978-1-4939-2522-3_17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
Some internal and external stimuli elicit stress responses on the cellular level and at the level of the organism. When the stimulus is brief and its intensity mild to moderate, it triggers adaptation changes that improve the cell's or organism's survival. This adaptation is achieved through a variety of cellular mechanisms such as induction of repair mechanisms, improved removal of damaged macromolecules, upregulation of endogenous antioxidant defenses, and prevention of apoptosis triggering by moderate stressors. The key intracellular signaling pathways involved in stress adaptation are the mTORC1 and SIRT1. Manipulating these stress adaptation signaling pathways with a variety of agents, improves the cellular adaptation to stress, prolongs cell survival, and improves the transplantation outcome in animal models and in clinical trials. The challenge for the future is to fine-tune the numerous experimental techniques to suit the needs of transplantation and regenerative medicine.
Collapse
Affiliation(s)
- Irina Milisav
- Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Zaloska 4, 1000, Ljubljana, Slovenia,
| | | | | |
Collapse
|
22
|
Galvao J, Davis B, Tilley M, Normando E, Duchen MR, Cordeiro MF. Unexpected low-dose toxicity of the universal solvent DMSO. FASEB J 2013; 28:1317-30. [PMID: 24327606 DOI: 10.1096/fj.13-235440] [Citation(s) in RCA: 450] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Dimethyl sulfoxide (DMSO) is an important aprotic solvent that can solubilize a wide variety of otherwise poorly soluble polar and nonpolar molecules. This, coupled with its apparent low toxicity at concentrations <10%, has led to its ubiquitous use and widespread application. Here, we demonstrate that DMSO induces retinal apoptosis in vivo at low concentrations (5 μl intravitreally dosed DMSO in rat from a stock concentration of 1, 2, 4, and 8% v/v). Toxicity was confirmed in vitro in a retinal neuronal cell line, at DMSO concentrations >1% (v/v), using annexin V, terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL), 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), and AlamarBlue cell viability assays. DMSO concentrations >10% (v/v) have recently been reported to cause cellular toxicity through plasma membrane pore formation. Here, we show the mechanism by which low concentrations (2-4% DMSO) induce caspase-3 independent neuronal death that involves apoptosis-inducing factor (AIF) translocation from mitochondria to the nucleus and poly-(ADP-ribose)-polymerase (PARP) activation. These results highlight safety concerns of using low concentrations of DMSO as a solvent for in vivo administration and in biological assays. We recommend that methods other than DMSO are employed for solubilizing drugs but, where no alternative exists, researchers compute absolute DMSO final concentrations and include an untreated control group in addition to DMSO vehicle control to check for solvent toxicity.
Collapse
Affiliation(s)
- Joana Galvao
- 1Glaucoma and Retinal Neurodegeneration Research Group, Institute of Ophthalmology, University College London, London EC1V 9EL, UK.
| | | | | | | | | | | |
Collapse
|
23
|
Nörenberg A, Heinz S, Scheller K, Hewitt NJ, Braspenning J, Ott M. Optimization of upcyte® human hepatocytes for the in vitro micronucleus assay. Mutat Res 2013; 758:69-79. [PMID: 24140631 DOI: 10.1016/j.mrgentox.2013.09.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Revised: 09/05/2013] [Accepted: 09/28/2013] [Indexed: 11/17/2022]
Abstract
"Upcyte(®) human hepatocytes" have the unique property of combining proliferation with the expression of drug metabolising activities. In our current study, we evaluated whether these cells would be suitable for early in vitro micronucleus (MN) tests. A treatment period of 96 h without a recovery period was most reliable for detecting MN formation in upcyte(®) hepatocytes from Donor 740. The basal MN rate in upcyte(®) hepatocytes varied considerably between donors (7-28%); therefore, modifications to the assay medium were tested to determine whether they could decrease inherent MN formation. Optimal medium supplements were 10 ng/ml oncostatin M for the pre-culture and recovery periods and 25 ng/ml epidermal growth factor and 10 ng/ml oncostatin M for the treatment period. Using the optimised conditions and outcome criteria, the upcyte(®) hepatocyte MN assay could correctly identify directly acting (e.g. mitomycin C, etoposide) and metabolically activated genotoxins (e.g. benzo[a]pyrene, cyclophosphamide). "True negative" and "false positive" compounds were also correctly identified as negative. The basal %MN in upcyte(®) hepatocytes from Donor 740 treated with DMSO, cyclophosphamide or MMC, was essentially unaffected by the growth stage ranging from population doublings of 14-61, suggesting that billions of cells could be produced from a single donor for standardised drug toxicity testing. In conclusion, we have established and optimised an in vitro MN test by using upcyte(®) hepatocytes to correctly identify known direct and metabolically activated genotoxicants as well as "false positives" and true negative compounds. The almost unlimited supply of cells from a single donor and optimised test conditions increase reproducibility in early and more predictive in vitro MN tests.
Collapse
|
24
|
Primary hepatocytes and their cultures in liver apoptosis research. Arch Toxicol 2013; 88:199-212. [PMID: 24013573 DOI: 10.1007/s00204-013-1123-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 08/29/2013] [Indexed: 01/18/2023]
Abstract
Apoptosis not only plays a key role in physiological demise of defunct hepatocytes, but is also associated with a plethora of acute and chronic liver diseases as well as with hepatotoxicity. The present paper focuses on the modelling of this mode of programmed cell death in primary hepatocyte cultures. Particular attention is paid to the activation of spontaneous apoptosis during the isolation of hepatocytes from the liver, its progressive manifestation upon the subsequent establishment of cell cultures and simultaneously to strategies to counteract this deleterious process. In addition, currently applied approaches to experimentally induce controlled apoptosis in this in vitro setting for mechanistic research purposes and thereby its detection using relevant biomarkers are reviewed.
Collapse
|
25
|
van Tonder JJ, Gulumian M, Cromarty AD, Steenkamp V. In vitro effect of N-acetylcysteine on hepatocyte injury caused by dichlorodiphenyltrichloroethane and its metabolites. Hum Exp Toxicol 2013; 33:41-53. [PMID: 23615707 DOI: 10.1177/0960327113482954] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The organochlorine pesticide, dichlorodiphenyltrichloroethane (DDT), is still used to combat the spread of malaria in several developing countries despite its accumulation and known hepatotoxic effects that have been demonstrated both in vitro and in vivo. N-Acetylcysteine (NAC) is a recognized hepatoprotective agent that has been reported to reduce hepatotoxicity initiated by many different compounds. The aim of this study was to determine whether NAC could counter in vitro hepatocyte injury induced by DDT or its two major metabolites, dichlorodiphenyldichloroethylene and dichlorodiphenyldichloroethane. HepG2 cell cultures were used to assess the following parameters of toxicity: cellular viability, intracellular levels of reactive oxygen species (ROS), mitochondrial membrane potential and initiation of apoptosis. None of the three test compounds induced ROS generation, yet exposure to any of the three compounds produced mitochondrial hyperpolarization, which was countered by NAC pretreatment. All three test compounds also induced apoptotic cell death, which was inhibited by NAC. Despite NAC counteracting some adverse intracellular changes due to organochlorine exposure, it appeared to aggravate the cytotoxic effects of the organochlorine compounds at low test concentrations. As the same outcome may also occur in vivo, results from the present study raise concern about the use of NAC as treatment for DDT-induced hepatotoxicity.
Collapse
Affiliation(s)
- J J van Tonder
- 1Department of Pharmacology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | | | | | | |
Collapse
|
26
|
A nanoformulation of siRNA and its role in cancer therapy: in vitro and in vivo evaluation. Cell Mol Biol Lett 2012; 18:120-36. [PMID: 23271435 PMCID: PMC6275763 DOI: 10.2478/s11658-012-0043-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2012] [Accepted: 12/15/2012] [Indexed: 11/20/2022] Open
Abstract
Overexpression of anti-apoptotic Bcl-2 is often observed in a wide variety of human cancers. It prevents the induction of apoptosis in neoplastic cells and contributes to resistance to chemotherapy. RNA interference has emerged as an efficient and selective technique for gene silencing. The potential to use small interfering RNA (siRNA) as a therapeutic agent for the treatment of cancer has elicited a great deal of interest. However, insufficient cellular uptake and poor stability have limited its therapeutic applications. The purpose of this study was to prepare chitosan nanoparticles via ionic gelation of chitosan by tripolyphosphate for effective delivery of siRNA to silence the anti-apoptotic Bcl-2 gene in neoplastic cells. Chitosan nanoparticles loaded with siRNA were in the size range 190 to 340 nm with a polydispersive index ranging from 0.04 to 0.2. They were able to completely bind with siRNA, provide protection against nuclease degradation, and enhance the transfection. Cell culture studies revealed that nanoparticles with entrapped siRNA could efficiently silence the antiapoptotic Bcl-2 gene. Studies on Swiss albino mice showed that siRNA could be effectively delivered through nanoparticles. There was significant decrease in the tumor volume. Blocking the expression of anti-apoptotic Bcl-2 can enhance the sensitivity of cancerous cells to anti-cancer drugs and the apoptosis rate. Therefore, nanoformulations with siRNA can be promoted as an adjuvant therapy in combination with anti-cancer drugs.
Collapse
|
27
|
Adaptive response, evidence of cross-resistance and its potential clinical use. Int J Mol Sci 2012; 13:10771-10806. [PMID: 23109822 PMCID: PMC3472714 DOI: 10.3390/ijms130910771] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Revised: 08/07/2012] [Accepted: 08/13/2012] [Indexed: 12/15/2022] Open
Abstract
Organisms and their cells are constantly exposed to environmental fluctuations. Among them are stressors, which can induce macromolecular damage that exceeds a set threshold, independent of the underlying cause. Stress responses are mechanisms used by organisms to adapt to and overcome stress stimuli. Different stressors or different intensities of stress trigger different cellular responses, namely induce cell repair mechanisms, induce cell responses that result in temporary adaptation to some stressors, induce autophagy or trigger cell death. Studies have reported life-prolonging effects of a wide variety of so-called stressors, such as oxidants, heat shock, some phytochemicals, ischemia, exercise and dietary energy restriction, hypergravity, etc. These stress responses, which result in enhanced defense and repair and even cross-resistance against multiple stressors, may have clinical use and will be discussed, while the emphasis will be on the effects/cross-effects of oxidants.
Collapse
|