1
|
Zoehler B, de Aguiar AM, Silveira GF. SAEDC: Development of a technological solution for exploratory data analysis and statistics in cytotoxicity. Comput Struct Biotechnol J 2024; 23:483-490. [PMID: 38261941 PMCID: PMC10796974 DOI: 10.1016/j.csbj.2023.12.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 01/25/2024] Open
Abstract
INTRODUCTION The intergovernmental organizations Organisation for Economic Co-operation and Development (OECD) and Interagency Coordinating Committee on the Validation of Alternative Methods (ICCVAM) have developed guidelines for the use of in vitro models for toxicological evaluation of chemicals. However, the presence of manual steps and the requirement of multiple tools for data analysis, apart from being costly and time-consuming, can inadvertently introduce errors by researchers. OBJECTIVES We have developed the SAEDC platform (Technological Solution for Exploratory Data Analysis and Statistics for Cytotoxicity, in Portuguese), which enables analysis of cytotoxicity data from assays following OECD Guideline No. 129. METHODOLOGY In vitro experimental data were used to compare with the analysis methodology suggested in the Guideline. We analyzed 117 data sets covering chemicals from Category I to Unclassified according to GHS classification. RESULTS The four-parameters of non-linear regression (4PL) calculated by the SAEDC platform showed no significant differences compared to standard methodology in any of the data sets (p > 0.05). The coefficient of determination (R-squared) also demonstrated not only a good fit of the 4PL model to the data but also significant similarity to values obtained by the conventional methodology. Finally, the SAEDC platform predicted LD50 values for the chemicals from IC50, using the Registry of Cytotoxicity (RC) regression models. CONCLUSION The comparison with the standard data analysis methodology revealed that SAEDC platform fulfills the requirements for cytotoxicity data analysis, generating reliable and accurate results with fewer steps performed by researchers. The use of SAEDC platform for obtaining toxicity values can reduce analysis time compared to the standard methodology proposed by regulatory agencies. Thus, automation of the analysis using the SAEDC platform has the potential to save time and resources for cytotoxicity researchers and laboratories while generating reliable results.
Collapse
Affiliation(s)
- Bernardo Zoehler
- Instituto Carlos Chagas – ICC, Fundação Oswaldo Cruz – Fiocruz, Brazil
| | - Alessandra Melo de Aguiar
- Plataforma de Bioensaios com métodos alternativos em citotoxicidade, Instituto Carlos Chagas – ICC, Fundação Oswaldo Cruz – Fiocruz, Brazil
- Laboratório de Biologia Básica de Células-tronco, Instituto Carlos Chagas – ICC, Fundação Oswaldo Cruz – Fiocruz, Brazil
| | | |
Collapse
|
2
|
Alfahlawy A, Selim MAA, Hassan HY. Effect of three different root canal sealants on human dental pulp stem cells. Sci Rep 2024; 14:23937. [PMID: 39397052 PMCID: PMC11471868 DOI: 10.1038/s41598-024-73232-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 09/16/2024] [Indexed: 10/15/2024] Open
Abstract
The cytotoxic effects of three root canal sealers with different bases on human dental pulp stem cells were assessed in this study using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) test. The cytotoxic effects of three root canal sealers with different bases on human dental pulp stem cells (DPSCs) were assessed in this study using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) test. The cytotoxicity of the sealers was tested after one, 4, and 7 d. Human dental pulp stem cell proliferation was concluded using an MTT assay. Cells not treated with sealer extract were used as controls. The absorption levels were measured using an Eliza spectrophotometer. P was set at 0.05 when the percentage of cell proliferation was matched between groups and observation times using one-way analysis of variance (ANOVA).During the second passage (P2), human dental pulp stem cells displayed a single morphological and phenotypic trait, with fibroblast morphology being the most common. There were no appreciable variations between the four groups after a day. There was a notable variation in the average percentage of cell proliferation between the groups after 4 and 7 days. The control group had the highest percentage, followed by the GuttaFlow Bioseal group, the Well Root St group, and the AH-Plus group, which had the lowest percentage. For every sealing group, after one day, the highest mean percentage of cell proliferation was recorded, followed by day four, and after day seven, the lowest mean percentage. The observation periods showed minimal cytotoxic effects of GuttaFlow Bioseal, whereas AH-Plus was the most cytotoxic to human dental pulp stem cells. The highest mean percentage of cell proliferation for all sealers was recorded on day one.
Collapse
Affiliation(s)
- Ahmad Alfahlawy
- Endodontic Department, Faculty of Dentistry, Suez Canal University, Ismailia, Egypt
| | - Manar A A Selim
- Oral Biology Department, Faculty of Dentistry, Suez Canal University, Ismailia, Egypt
| | - Hayam Y Hassan
- Endodontic Department, Faculty of Dentistry, Suez Canal University, Ismailia, Egypt.
| |
Collapse
|
3
|
Thongsri O, Thaitalay P, Srisuwan S, Khophai S, Suksaweang S, Rojviriya C, Panpisutd P, Patntirapong S, Gough J, Rattanachan ST. Enhanced remineralisation ability and antibacterial properties of sol-gel glass ionomer cement modified by fluoride containing strontium-based bioactive glass or strontium-containing fluorapatite. Dent Mater 2024; 40:716-727. [PMID: 38395738 DOI: 10.1016/j.dental.2024.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 01/19/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024]
Abstract
OBJECTIVES This study aimed to compare two types of bioactive additives which were strontium-containing fluorinated bioactive glass (SrBGF) or strontium-containing fluorapatite (SrFA) added to sol-gel derived glass ionomer cement (SGIC). The objective was to develop antibacterial and mineralisation properties, using bioactive additives, to minimize the occurrence of caries lesions in caries disease. METHODS Synthesized SrBGF and SrFA nanoparticles were added to SGIC at 1 wt% concentration to improve antibacterial properties against S. mutans, promote remineralisation, and hASCs and hDPSCs viability. Surface roughness and ion-releasing behavior were also evaluated to clarify the effect on the materials. Antibacterial activity was measured via agar disc diffusion and bacterial adhesion. Remineralisation ability was assessed by applying the material to demineralised teeth and subjecting them to a 14-day pH cycle, followed by microCT and SEM-EDS analysis. RESULTS The addition of SrFA into SGIC significantly improved its antibacterial property. SGIC modified with either SrBGF or SrFA additives could similarly induce apatite crystal precipitation onto demineralised dentin and increase dentin density, indicating its ability to remineralise dentin. Moreover, this study also showed that SGIC modified with SrBGF or SrFA additives had promising results on the in vitro cytotoxicity of hASC and hDPSC. SIGNIFICANT SrFA has superior antibacterial property as compared to SrBGF while demonstrating equal remineralisation ability. Furthermore, the modified SGIC showed promising results in reducing the cytotoxicity of hASCs and hDPSCs, indicating its potential for managing caries.
Collapse
Affiliation(s)
- Oranich Thongsri
- School of Ceramic Engineering, Institute of Engineering, Suranaree University of Technology, 111 University Avenue, Muang District, Nakhon Ratchasima 30000, Thailand
| | - Paritat Thaitalay
- School of Ceramic Engineering, Institute of Engineering, Suranaree University of Technology, 111 University Avenue, Muang District, Nakhon Ratchasima 30000, Thailand
| | - Sawitri Srisuwan
- School of Ceramic Engineering, Institute of Engineering, Suranaree University of Technology, 111 University Avenue, Muang District, Nakhon Ratchasima 30000, Thailand
| | - Sasikamon Khophai
- School of Chemistry, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand
| | - Sanong Suksaweang
- School of Pathology and Laboratory Medicine, Institute of Medicine, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand
| | - Catleya Rojviriya
- Synchrotron Light Research Institute (Public Organization), Nakhon Ratchasima 30000, Thailand
| | - Piyaphong Panpisutd
- Faculty of Dentistry, Thammasat University, Pathum Thani 12120, Thailand; Thammasat University Research Unit in Dental and Bone Substitute Biomaterials, Thammasat University, Pathum Thani 12120, Thailand
| | - Somying Patntirapong
- Faculty of Dentistry, Thammasat University, Pathum Thani 12120, Thailand; Thammasat University Research Unit in Dental and Bone Substitute Biomaterials, Thammasat University, Pathum Thani 12120, Thailand
| | - Julie Gough
- Department of Materials and Henry Royce Institute, The University of Manchester, Manchester M13 9PL, UK
| | - Sirirat Tubsungnoen Rattanachan
- School of Ceramic Engineering, Institute of Engineering, Suranaree University of Technology, 111 University Avenue, Muang District, Nakhon Ratchasima 30000, Thailand.
| |
Collapse
|
4
|
He Y, Shi F, Hu J, Li H, Chen X, Yuan L, Lu Y, Du W, Li R, Wu J, Deng F, Yu D. Magnetic graphene oxide nanocomposites induce cytotoxicity in ADSCs via GPX4 regulating ferroptosis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 269:115745. [PMID: 38029583 DOI: 10.1016/j.ecoenv.2023.115745] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 10/30/2023] [Accepted: 11/24/2023] [Indexed: 12/01/2023]
Abstract
Magnetic graphene oxide nanocomposites (MGO NPs) have been widely studied in biomedical applications. However, their cytotoxicity and underlying mechanisms remain unclear. In this study, the biosafety of MGO NPs was investigated, and the mechanism involved in ferroptosis was further explored. MGO can produce cytotoxicity in ADSCs, which is dependent on their concentration. Ferroptosis was involved in MGO NP-induced ADSC survival inhibition by increasing total ROS and lipid ROS accumulation as well as regulating the expression levels of ferroptosis-related genes and proteins. GPX4 played a critical role in the MGO NP-induced ADSC ferroptosis process, and overexpressing GPX4 suppressed ferroptosis to increase cell survival. This study provides a theoretical basis for the biosafety management of MGO NPs used in the field of biomedical treatment.
Collapse
Affiliation(s)
- Yi He
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Fangyang Shi
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Jiajun Hu
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Hongyu Li
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Xun Chen
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Lingyu Yuan
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Yunyang Lu
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Weidong Du
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Runze Li
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Jie Wu
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Feilong Deng
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China.
| | - Dongsheng Yu
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
5
|
Correia CD, Ferreira A, Fernandes MT, Silva BM, Esteves F, Leitão HS, Bragança J, Calado SM. Human Stem Cells for Cardiac Disease Modeling and Preclinical and Clinical Applications—Are We on the Road to Success? Cells 2023; 12:1727. [DOI: https:/doi.org/10.3390/cells12131727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023] Open
Abstract
Cardiovascular diseases (CVDs) are pointed out by the World Health Organization (WHO) as the leading cause of death, contributing to a significant and growing global health and economic burden. Despite advancements in clinical approaches, there is a critical need for innovative cardiovascular treatments to improve patient outcomes. Therapies based on adult stem cells (ASCs) and embryonic stem cells (ESCs) have emerged as promising strategies to regenerate damaged cardiac tissue and restore cardiac function. Moreover, the generation of human induced pluripotent stem cells (iPSCs) from somatic cells has opened new avenues for disease modeling, drug discovery, and regenerative medicine applications, with fewer ethical concerns than those associated with ESCs. Herein, we provide a state-of-the-art review on the application of human pluripotent stem cells in CVD research and clinics. We describe the types and sources of stem cells that have been tested in preclinical and clinical trials for the treatment of CVDs as well as the applications of pluripotent stem-cell-derived in vitro systems to mimic disease phenotypes. How human stem-cell-based in vitro systems can overcome the limitations of current toxicological studies is also discussed. Finally, the current state of clinical trials involving stem-cell-based approaches to treat CVDs are presented, and the strengths and weaknesses are critically discussed to assess whether researchers and clinicians are getting closer to success.
Collapse
Affiliation(s)
- Cátia D. Correia
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| | - Anita Ferreira
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| | - Mónica T. Fernandes
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- School of Health, Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| | - Bárbara M. Silva
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Doctoral Program in Biomedical Sciences, Faculty of Medicine and Biomedical Sciences, Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| | - Filipa Esteves
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| | - Helena S. Leitão
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences, Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| | - José Bragança
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences, Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Champalimaud Research Program, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal
| | - Sofia M. Calado
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences, Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| |
Collapse
|
6
|
Correia CD, Ferreira A, Fernandes MT, Silva BM, Esteves F, Leitão HS, Bragança J, Calado SM. Human Stem Cells for Cardiac Disease Modeling and Preclinical and Clinical Applications-Are We on the Road to Success? Cells 2023; 12:1727. [PMID: 37443761 PMCID: PMC10341347 DOI: 10.3390/cells12131727] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 06/08/2023] [Accepted: 06/15/2023] [Indexed: 07/15/2023] Open
Abstract
Cardiovascular diseases (CVDs) are pointed out by the World Health Organization (WHO) as the leading cause of death, contributing to a significant and growing global health and economic burden. Despite advancements in clinical approaches, there is a critical need for innovative cardiovascular treatments to improve patient outcomes. Therapies based on adult stem cells (ASCs) and embryonic stem cells (ESCs) have emerged as promising strategies to regenerate damaged cardiac tissue and restore cardiac function. Moreover, the generation of human induced pluripotent stem cells (iPSCs) from somatic cells has opened new avenues for disease modeling, drug discovery, and regenerative medicine applications, with fewer ethical concerns than those associated with ESCs. Herein, we provide a state-of-the-art review on the application of human pluripotent stem cells in CVD research and clinics. We describe the types and sources of stem cells that have been tested in preclinical and clinical trials for the treatment of CVDs as well as the applications of pluripotent stem-cell-derived in vitro systems to mimic disease phenotypes. How human stem-cell-based in vitro systems can overcome the limitations of current toxicological studies is also discussed. Finally, the current state of clinical trials involving stem-cell-based approaches to treat CVDs are presented, and the strengths and weaknesses are critically discussed to assess whether researchers and clinicians are getting closer to success.
Collapse
Affiliation(s)
- Cátia D. Correia
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal; (C.D.C.); (A.F.); (M.T.F.); (B.M.S.); (F.E.); (H.S.L.); (J.B.)
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| | - Anita Ferreira
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal; (C.D.C.); (A.F.); (M.T.F.); (B.M.S.); (F.E.); (H.S.L.); (J.B.)
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| | - Mónica T. Fernandes
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal; (C.D.C.); (A.F.); (M.T.F.); (B.M.S.); (F.E.); (H.S.L.); (J.B.)
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- School of Health, Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| | - Bárbara M. Silva
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal; (C.D.C.); (A.F.); (M.T.F.); (B.M.S.); (F.E.); (H.S.L.); (J.B.)
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Doctoral Program in Biomedical Sciences, Faculty of Medicine and Biomedical Sciences, Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| | - Filipa Esteves
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal; (C.D.C.); (A.F.); (M.T.F.); (B.M.S.); (F.E.); (H.S.L.); (J.B.)
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| | - Helena S. Leitão
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal; (C.D.C.); (A.F.); (M.T.F.); (B.M.S.); (F.E.); (H.S.L.); (J.B.)
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences, Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| | - José Bragança
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal; (C.D.C.); (A.F.); (M.T.F.); (B.M.S.); (F.E.); (H.S.L.); (J.B.)
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences, Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Champalimaud Research Program, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal
| | - Sofia M. Calado
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal; (C.D.C.); (A.F.); (M.T.F.); (B.M.S.); (F.E.); (H.S.L.); (J.B.)
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences, Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| |
Collapse
|
7
|
Lino JB, Robert AW, Stimamiglio MA, de Aguiar AM. Comparative analysis of the potential of the secretomes of cardiac resident stromal cells and fibroblasts. IUBMB Life 2023; 75:196-206. [PMID: 34590780 DOI: 10.1002/iub.2557] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/13/2021] [Indexed: 11/10/2022]
Abstract
The secretome of different cell types has been applied on in vitro and in vivo assays, indicating considerable therapeutic potential. However, the choice of the ideal cell type and culture conditions for obtaining the best set of soluble factors, as well as the assays to assess specific effects, remain subjects of vigorous debate. In this study, we used mass spectrometry to characterize the secretomes of ventricle derived-cardiac resident stromal cells (vCRSC) and human dermal fibroblasts (HDFs) and evaluate them in an effort to understand the niche specificity of biological responses toward different cellular behaviors, such as cell proliferation, adhesion, migration, and differentiation. It was interesting to note that the HDF and vCRSC secretomes were both able to induce proliferation and cardiac differentiation of H9c2 cells, as well as to increase the adhesion activity of H9c2 cells and human umbilical vein endothelial cells. Analysis of the secretome composition showed that the vCRSCs derived from different donors secreted a similar set of proteins. Despite the differences, almost half of the proteins identified in conditioned medium were common to both HDF and vCRSC. Consequently, a high number of common biological processes were identified in the secretomes of the two cell types, which could help to explain the similar results observed in the in vitro assays. We show that soluble factors secreted by both HDF and vCRSC are able to promote proliferation and differentiation of cardiomyoblasts in vitro. Our study indicates the possible use of vCRSC or HDF secretomes in acellular therapies for regenerative medicine.
Collapse
Affiliation(s)
- Jhonatan Basso Lino
- Stem Cells Basic Biology Laboratory, Carlos Chagas Institute - FIOCRUZ/PR, Curitiba, Paraná, Brazil
| | - Anny Waloski Robert
- Stem Cells Basic Biology Laboratory, Carlos Chagas Institute - FIOCRUZ/PR, Curitiba, Paraná, Brazil
| | | | | |
Collapse
|
8
|
Vieira MPS, Silva OBS, Souza GF, Cavalcante GTS, Souza FMA, Gitaí DLG, Castro OW, Nicácio DCSP, Cofré AHR, Amorós MA, Silva AV, Neto GJDS, Silva AHQ, Correia WBZGB, Junkes JA, Duarte FS, Guedes JS, Nogueira FCS, Meneghetti MR, Duzzioni M. First evaluation of the anxiolytic-like effects of a bromazepam‑palladium complex in mice. J Inorg Biochem 2022; 237:112012. [PMID: 36162209 DOI: 10.1016/j.jinorgbio.2022.112012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 09/09/2022] [Accepted: 09/16/2022] [Indexed: 01/18/2023]
Abstract
A significant fraction of patients are affected by persistent fear and anxiety. Currently, there are several anxiolytic drug options, however their clinical outcomes do not fully manage the symptoms. Here, we evaluated the effects of a bromazepam‑palladium derivative [2-{(7-bromo-2-oxo-1,3-dihydro-2H-1,4-benzodiazepin-5-il)pyridinyl-κ2-N,N}chloropalladium(II)], [(BMZ)PdCl2], on fear/anxiety and memory-related behavior in mice. For this, female Swiss mice were treated intraperitoneally (i.p.) with saline (NaCl 0.9%) or [(BMZ)PdCl2] (0.5, 5.0, or 50 μg/kg). After 30 min, different tests were performed to evaluate anxiety, locomotion, and memory. We also evaluated the acute toxicity of [(BMZ)PdCl2] using a cell viability assay (neutral red uptake assay), and whether the drugs mechanism of action involves the γ-aminobutyric acid type A (GABAA) receptor complex by pre-treating animals with flumazenil (1.0 mg/kg, i.p., a competitive antagonist of GABAA-binding site). Our results demonstrate that [(BMZ)PdCl2] induces an anxiolytic-like phenotype in the elevated plus-maze test and that this effect can be blocked by flumazenil. Furthermore, there were no behavioral alterations induced by [(BMZ)PdCl2], as evaluated in the light-dark box, open field, and step-down passive avoidance tests. In the acute toxicity assay, [(BMZ)PdCl2] presented IC50 and LD50 values of 218 ± 60 μg/mL and 780 ± 80 mg/kg, respectively, and GSH category 4. Taken together, our results show that the anxiolytic-like effect of acute treatment with [(BMZ)PdCl2] occurs through the modulation of the benzodiazepine site in the GABAA receptor complex. Moreover, we show indications that [(BMZ)PdCl2] does not promote sedation and amnesia and presents the same toxicity as the bromazepam prototype.
Collapse
Affiliation(s)
- Mirella P S Vieira
- Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas, Avenida Lourival Melo Mota, S/N, Cidade Universitária, Maceió, Alagoas 57072-900, Brazil
| | - Ozileudiane B S Silva
- Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas, Avenida Lourival Melo Mota, S/N, Cidade Universitária, Maceió, Alagoas 57072-900, Brazil
| | - Gabriela F Souza
- Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas, Avenida Lourival Melo Mota, S/N, Cidade Universitária, Maceió, Alagoas 57072-900, Brazil
| | - Gabriela T S Cavalcante
- Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas, Avenida Lourival Melo Mota, S/N, Cidade Universitária, Maceió, Alagoas 57072-900, Brazil
| | - Fernanda M A Souza
- Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas, Avenida Lourival Melo Mota, S/N, Cidade Universitária, Maceió, Alagoas 57072-900, Brazil
| | - Daniel L G Gitaí
- Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas, Avenida Lourival Melo Mota, S/N, Cidade Universitária, Maceió, Alagoas 57072-900, Brazil
| | - Olagide W Castro
- Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas, Avenida Lourival Melo Mota, S/N, Cidade Universitária, Maceió, Alagoas 57072-900, Brazil
| | - Dannyele C S P Nicácio
- Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas, Avenida Lourival Melo Mota, S/N, Cidade Universitária, Maceió, Alagoas 57072-900, Brazil
| | - Axel H R Cofré
- Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas, Avenida Lourival Melo Mota, S/N, Cidade Universitária, Maceió, Alagoas 57072-900, Brazil
| | - Mariana A Amorós
- Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas, Avenida Lourival Melo Mota, S/N, Cidade Universitária, Maceió, Alagoas 57072-900, Brazil
| | - Artur V Silva
- Grupo de Catálise e Reatividade Química, Instituto de Química e Biotecnologia, Universidade Federal de Alagoas, Avenida Lourival Melo Mota, S/N, Cidade Universitária, Maceió, Alagoas 57072-900, Brazil
| | - Geraldo José da Silva Neto
- Grupo de Catálise e Reatividade Química, Instituto de Química e Biotecnologia, Universidade Federal de Alagoas, Avenida Lourival Melo Mota, S/N, Cidade Universitária, Maceió, Alagoas 57072-900, Brazil
| | - Allysson H Q Silva
- Grupo de Catálise e Reatividade Química, Instituto de Química e Biotecnologia, Universidade Federal de Alagoas, Avenida Lourival Melo Mota, S/N, Cidade Universitária, Maceió, Alagoas 57072-900, Brazil
| | - Walleska B Z G B Correia
- Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas, Avenida Lourival Melo Mota, S/N, Cidade Universitária, Maceió, Alagoas 57072-900, Brazil; Grupo de Catálise e Reatividade Química, Instituto de Química e Biotecnologia, Universidade Federal de Alagoas, Avenida Lourival Melo Mota, S/N, Cidade Universitária, Maceió, Alagoas 57072-900, Brazil
| | - Janaína A Junkes
- Instituto de Tecnologia e Pesquisa, Centro Universitário Tiradentes, Avenida Comendador Gustavo Paiva, 5017, Cruz das Almas, Maceió, Alagoas 57038-000, Brazil
| | - Filipe S Duarte
- Departamento de Fisiologia e Farmacologia, Universidade Federal de Pernambuco, Avenida Professor Moraes Rego, S/N, Cidade Universitária, Recife, Pernambuco 50670-420, Brazil
| | - Jéssica S Guedes
- Instituto de Química, Universidade Federal do Rio de Janeiro, Avenida Athos da Silveira Ramos, 149, Cidade Universitária, Rio de Janeiro, Rio de Janeiro 21941-909, Brazil
| | - Fábio C S Nogueira
- Instituto de Química, Universidade Federal do Rio de Janeiro, Avenida Athos da Silveira Ramos, 149, Cidade Universitária, Rio de Janeiro, Rio de Janeiro 21941-909, Brazil
| | - Mario R Meneghetti
- Grupo de Catálise e Reatividade Química, Instituto de Química e Biotecnologia, Universidade Federal de Alagoas, Avenida Lourival Melo Mota, S/N, Cidade Universitária, Maceió, Alagoas 57072-900, Brazil.
| | - Marcelo Duzzioni
- Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas, Avenida Lourival Melo Mota, S/N, Cidade Universitária, Maceió, Alagoas 57072-900, Brazil.
| |
Collapse
|
9
|
Valle ACV, Brunel HDSS, Dallago BSL, Rodrigues LS, Malard PF, da Costa RA, Rossetto R, de Andrade RV. In-Vitro Growth Kinetics of Mesenchymal Stem Cells in Cytotoxicity Tests Using Low-Diluted Viscum Album. HOMEOPATHY 2022; 112:40-49. [PMID: 35988582 PMCID: PMC9868971 DOI: 10.1055/s-0042-1747682] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
INTRODUCTION The use of mesenchymal stem cells (MSC) in cytotoxicity tests is an in-vitro alternative model for predicting initial doses. Homeopathic medicines may stimulate the immune system to combat a pathology effectively and have been used for over two centuries. Viscum album (VA) extracts are widely used in the treatment of cancer, due to their immunomodulatory, cytotoxic and pro-apoptotic properties. OBJECTIVE This study aimed to evaluate the in-vitro growth kinetics of canine MSC in relation to cytotoxicity, cell differentiation and expression of pluripotentiality markers, using a VA preparation at the D1D2 (1×10-1, 1×10-2 potency (VAD1D2). METHODS MSC were obtained from adipose tissue sampled from a healthy dog that was undergoing an elective veterinary procedure and with its owner's permission. The experiments were performed in three groups: MSC treated with VAD1D2 or diluent or untreated (control). The cytotoxicity was evaluated by MTT assay. The differentiation was induced in three lineages, and apoptotic cell labeling was performed by an Annexin-V test. RESULTS At the concentration of 10 μL/mL of VA, the number of cells after in-vitro culture was maintained when compared with the control (untreated) group. A significant and gradual decrease in cell viability was recorded as VA concentrations increased. The apoptosis analysis showed that VA at 20 μL/mL presented absolute percentages of initial apoptosis twice as high as at 10 μL/mL, which was similar to the control (untreated group). CONCLUSION The results suggest that the use of efficient methods to assess the in-vitro cytotoxicity of VA-based homeopathic medicines using MSC lineages may predict the potential action at different concentrations. These findings demonstrated that VAD1D2 interferes with canine MSC growth kinetics.
Collapse
Affiliation(s)
- Ana Catarina Viana Valle
- Doctor Izao Soares Institute, Sao Paulo, Brazil,Graduate Program in Genomic Sciences and Biotechnology, Catholic University of Brasilia, Brazil
| | | | | | | | - Patrícia Furtado Malard
- Graduate Program in Genomic Sciences and Biotechnology, Catholic University of Brasilia, Brazil,BioCell Cell Therapy, Brazil
| | | | - Rafael Rossetto
- Graduate Program in Genomic Sciences and Biotechnology, Catholic University of Brasilia, Brazil
| | - Rosângela Vieira de Andrade
- Graduate Program in Genomic Sciences and Biotechnology, Catholic University of Brasilia, Brazil,Address for correspondence Rosangela Vieira de Andrade, PhD GGraduate Program in Genomic Sciences and Biotechnology, Catholic University of BrasiliaSGAN 916, Brasília, DF 70790-160Brazil
| |
Collapse
|
10
|
Biological Synthesis of Low Cytotoxicity Silver Nanoparticles (AgNPs) by the Fungus Chaetomium thermophilum—Sustainable Nanotechnology. J Fungi (Basel) 2022; 8:jof8060605. [PMID: 35736088 PMCID: PMC9224622 DOI: 10.3390/jof8060605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 06/02/2022] [Accepted: 06/02/2022] [Indexed: 12/10/2022] Open
Abstract
Fungal biotechnology research has rapidly increased as a result of the growing awareness of sustainable development and the pressing need to explore eco-friendly options. In the nanotechnology field, silver nanoparticles (AgNPs) are currently being studied for application in cancer therapy, tumour detection, drug delivery, and elsewhere. Therefore, synthesising nanoparticles (NPs) with low toxicity has become essential in the biomedical area. The fungus Chaetomium thermophilum (C. thermophilum) was here investigated—to the best of our knowledge, for the first time—for application in the production of AgNPs. Transmission electronic microscopy (TEM) images demonstrated a spherical AgNP shape, with an average size of 8.93 nm. Energy-dispersive X-ray spectrometry (EDX) confirmed the presence of elemental silver. A neutral red uptake (NRU) test evaluated the cytotoxicity of the AgNPs at different inhibitory concentrations (ICs). A half-maximal concentration (IC50 = 119.69 µg/mL) was used to predict a half-maximal lethal dose (LD50 = 624.31 mg/kg), indicating a Global Harmonized System of Classification and Labelling of Chemicals (GHS) acute toxicity estimate (ATE) classification category of 4. The fungus extract showed a non-toxic profile at the IC tested. Additionally, the interaction between the AgNPs and the Balb/c 3T3 NIH cells at an ultrastructural level resulted in preserved cells structures at non-toxic concentrations (IC20 = 91.77 µg/mL), demonstrating their potential as sustainable substitutes for physical and chemically made AgNPs. Nonetheless, at the IC50, the cytoplasm of the cells was damaged and mitochondrial morphological alteration was evident. This fact highlights the fact that dose-dependent phenomena are involved, as well as emphasising the importance of investigating NPs’ effects on mitochondria, as disruption to this organelle can impact health.
Collapse
|
11
|
Suitability of Human Mesenchymal Stem Cells Derived from Fetal Umbilical Cord (Wharton’s Jelly) as an Alternative In Vitro Model for Acute Drug Toxicity Screening. Cells 2022; 11:cells11071102. [PMID: 35406666 PMCID: PMC8997545 DOI: 10.3390/cells11071102] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/14/2022] [Accepted: 03/22/2022] [Indexed: 02/01/2023] Open
Abstract
Preclinical toxicity screening is the first and most crucial test that assesses the safety of new candidate drugs before their consideration for further evaluation in clinical trials. In vitro drug screening using stem cells has lately arisen as a promising alternative to the “gold standard” of animal testing, but their suitability and performance characteristics in toxicological studies have so far not been comprehensively investigated. In this study, we focused on the evaluation of human mesenchymal stem cells isolated from the matrix (Wharton’s jelly) of fetal umbilical cord (WJSCs), which bear enhanced in vitro applicability due to their unique biological characteristics. In order to determine their suitability for drug-related cytotoxicity assessment, we adopted a high-throughput methodology that evaluated their sensitivity to a selected panel of chemicals in different culture environments. Cytotoxicity was measured within 48 h by means of MTS and/or NRU viability assays, and was compared directly (in vitro) or indirectly (in silico) to adult human mesenchymal stem cells and to reference cell lines of human and murine origin. Our data clearly suggest that human WJSCs can serve as a robust in vitro alternative for acute drug toxicity screening by uniquely combining rapid and versatile assay setup with high-throughput analysis, good representation of human toxicology, high reproducibility, and low cost.
Collapse
|
12
|
Abud APR, Paschoal ACC, Kuligovski C, Caruso RRB, Dallagiovanna B, de Aguiar AM. Using inhibition of the adipogenesis of adipose-derived stem cells in vitro for toxicity prediction. MethodsX 2021; 8:101515. [PMID: 34754786 PMCID: PMC8564732 DOI: 10.1016/j.mex.2021.101515] [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: 07/02/2021] [Accepted: 09/13/2021] [Indexed: 11/24/2022] Open
Abstract
In vitro stem cell models are used as alternatives to animal models and are important tools for cytotoxicity studies. Researchers can determine the effects of test substances on human cells by evaluating cell viability and differentiation. Here, we describe an in vitro model to quantify adipogenesis based on the Nile red staining of specific lipid droplets and the emission of basic lipids from human adipose tissue-derived mesenchymal stromal cells (AD-MSCs) in the presence of test substances. This assay allows for the prediction of toxicity based on the inhibition of adipogenesis in vitro in a 96-well format. The differentiation of a progenitor cell into a specialized cell, the adipocyte, is easy to monitor and quantify, making this a simple assay. The fluorescence staining of nuclei and lipid droplets is measured after 14 days of cell differentiation to determine cell number and assess cell differentiation using high-content imaging analysis, thus allowing for the identification of chemicals that impact differentiation. We also describe a protocol to assess adipocyte differentiation by fluorescence intensity using a multiplate reader.Researchers can utilize the protocol described here for many purposes to evaluate in vitro adipogenesis. With this method, it is possible to reduce the use of animals.
Collapse
Affiliation(s)
- Ana Paula Ressetti Abud
- Rede de Plataformas Tecnológicas FIOCRUZ - Bioensaios com Métodos alternativos em Citotoxicidade, Rua Professor Algacyr Munhoz Mader, 3775, Instituto Carlos Chagas, FIOCRUZ Paraná, Curitiba, PR 81350-010, Brazil
| | - Ariane Caroline Campos Paschoal
- Laboratório de Biologia Básica de Células-Tronco, Rua Professor Algacyr Munhoz Mader, 3775, Instituto Carlos Chagas, FIOCRUZ Paraná, Curitiba, PR 81350-010, Brazil.,Grupo Boticário, Pesquisa and Desenvolvimento, Avenida Rui Barbosa, 4110, São José dos Pinhais, PR 83055-320, Brazil
| | - Crisciele Kuligovski
- Laboratório de Biologia Básica de Células-Tronco, Rua Professor Algacyr Munhoz Mader, 3775, Instituto Carlos Chagas, FIOCRUZ Paraná, Curitiba, PR 81350-010, Brazil
| | - Rodrigo Rêgo Barros Caruso
- Laboratório de Ciências e Tecnologias Aplicadas à Saúde, Rua Professor Algacyr Munhoz Mader, 3775, Instituto Carlos Chagas, FIOCRUZ Paraná, Curitiba, PR 81350-010, Brazil.,Current Address: Laboratório de Biologia Molecular e Sistêmica de Tripanossomatídeos. Rua Professor Algacyr Munhoz Mader, 3775, Instituto Carlos Chagas, FIOCRUZ Paraná, Curitiba, PR 81350-010, Brazil
| | - Bruno Dallagiovanna
- Laboratório de Biologia Básica de Células-Tronco, Rua Professor Algacyr Munhoz Mader, 3775, Instituto Carlos Chagas, FIOCRUZ Paraná, Curitiba, PR 81350-010, Brazil
| | - Alessandra Melo de Aguiar
- Rede de Plataformas Tecnológicas FIOCRUZ - Bioensaios com Métodos alternativos em Citotoxicidade, Rua Professor Algacyr Munhoz Mader, 3775, Instituto Carlos Chagas, FIOCRUZ Paraná, Curitiba, PR 81350-010, Brazil.,Laboratório de Biologia Básica de Células-Tronco, Rua Professor Algacyr Munhoz Mader, 3775, Instituto Carlos Chagas, FIOCRUZ Paraná, Curitiba, PR 81350-010, Brazil
| |
Collapse
|
13
|
Biagini G, Senegaglia AC, Pereira T, Berti LF, Marcon BH, Stimamiglio MA. 3D Poly(Lactic Acid) Scaffolds Promote Different Behaviors on Endothelial Progenitors and Adipose-Derived Stromal Cells in Comparison With Standard 2D Cultures. Front Bioeng Biotechnol 2021; 9:700862. [PMID: 34568295 PMCID: PMC8455839 DOI: 10.3389/fbioe.2021.700862] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 08/23/2021] [Indexed: 12/13/2022] Open
Abstract
Tissue engineering is a branch of regenerative medicine, which comprises the combination of biomaterials, cells and other bioactive molecules to regenerate tissues. Biomaterial scaffolds act as substrate and as physical support for cells and they can also reproduce the extracellular matrix cues. Although tissue engineering applications in cellular therapy tend to focus on the use of specialized cells from particular tissues or stem cells, little attention has been paid to endothelial progenitors, an important cell type in tissue regeneration. We combined 3D printed poly(lactic acid) scaffolds comprising two different pore sizes with human adipose-derived stromal cells (hASCs) and expanded CD133+ cells to evaluate how these two cell types respond to the different architectures. hASCs represent an ideal source of cells for tissue engineering applications due to their low immunogenicity, paracrine activity and ability to differentiate. Expanded CD133+ cells were isolated from umbilical cord blood and represent a source of endothelial-like cells with angiogenic potential. Fluorescence microscopy and scanning electron microscopy showed that both cell types were able to adhere to the scaffolds and maintain their characteristic morphologies. The porous PLA scaffolds stimulated cell cycle progression of hASCs but led to an arrest in the G1 phase and reduced proliferation of expanded CD133+ cells. Also, while hASCs maintained their undifferentiated profile after 7 days of culture on the scaffolds, expanded CD133+ cells presented a reduction of the von Willebrand factor (vWF), which affected the cells’ angiogenic potential. We did not observe changes in cell behavior for any of the parameters analyzed between the scaffolds with different pore sizes, but the 3D environment created by the scaffolds had different effects on the cell types tested. Unlike the extensively used mesenchymal stem cell types, the 3D PLA scaffolds led to opposite behaviors of the expanded CD133+ cells in terms of cytotoxicity, proliferation and immunophenotype. The results obtained reinforce the importance of studying how different cell types respond to 3D culture systems when considering the scaffold approach for tissue engineering.
Collapse
Affiliation(s)
- Giuliana Biagini
- Laboratório de Biologia Básica de Células-Tronco, Instituto Carlos Chagas, Fiocruz Paraná, Curitiba, Brazil
| | | | - Tarciso Pereira
- Department of Mechanical Engineering, Post Graduate Program in Biomedical Engineering, Universidade Tecnológica Federal do Paraná, Curitiba, Brazil
| | - Lucas Freitas Berti
- Department of Mechanical Engineering, Post Graduate Program in Biomedical Engineering, Universidade Tecnológica Federal do Paraná, Curitiba, Brazil
| | - Bruna Hilzendeger Marcon
- Laboratório de Biologia Básica de Células-Tronco, Instituto Carlos Chagas, Fiocruz Paraná, Curitiba, Brazil
| | - Marco Augusto Stimamiglio
- Laboratório de Biologia Básica de Células-Tronco, Instituto Carlos Chagas, Fiocruz Paraná, Curitiba, Brazil
| |
Collapse
|
14
|
Ribeiro AL, Bassai LW, Robert AW, Machado TN, Bezerra AG, Horinouchi CDDS, Aguiar AMD. Bismuth-based nanoparticles impair adipogenic differentiation of human adipose-derived mesenchymal stem cells. Toxicol In Vitro 2021; 77:105248. [PMID: 34560244 DOI: 10.1016/j.tiv.2021.105248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 09/06/2021] [Accepted: 09/16/2021] [Indexed: 11/27/2022]
Abstract
Bismuth-based nanoparticles (BiNPs) have attracted attention for their potential biomedical applications. However, there is a lack of information concerning their interaction with biological systems. In this study, it was investigated the effect of physically synthesized BiNPs to human adipose-derived stem cells (ADSCs). We first evaluated the influence of BiNPs on cell viability, cell morphology, mitochondrial function and cell proliferation. Further, the impact of BiNPs on adipogenic differentiation was also explored. Cytotoxicity assays have demonstrated that BiNPs did not reduce relative cell viability of ADSC except at the highest tested concentration (345 μg/ml). Analysis of cell morphology performed by transmission electron microscopy confirmed that BiNPs induced cell damage only at a high concentration (302.24 μg/ml), equivalent to IC50 concentration. Moreover, BiNPs exposure increased the expression of the cell proliferation marker Ki-67 and the incorporation of the thymidine analogue EdU into cell DNA, suggesting that these nanoparticles could be stimulating ADSC proliferation. BiNPs also increased the mitochondrial membrane potential. Furthermore, BiNPs reduced ADSC adipogenic differentiation as measured by lipid droplet accumulation and mRNA expression levels of the specific adipogenesis biomarkers PPARγ, C/EPBɑ and FABP4. Thus, BiNPs affect the nonspecific (viability, proliferation and mitochondrial activity) and specific (adipogenesis) cellular mechanisms of ADSCs.
Collapse
Affiliation(s)
- Annanda Lyra Ribeiro
- Laboratório de Biologia Básica de Células-Tronco, Instituto Carlos Chagas, FIOCRUZ Paraná, Curitiba, Paraná, Brazil
| | - Letícia Werzel Bassai
- Laboratório de Biologia Básica de Células-Tronco, Instituto Carlos Chagas, FIOCRUZ Paraná, Curitiba, Paraná, Brazil; Laboratório de Cultivo de Eucariotos, Instituto de Biologia Molecular do Paraná, Curitiba, Paraná, Brazil
| | - Anny Waloski Robert
- Laboratório de Biologia Básica de Células-Tronco, Instituto Carlos Chagas, FIOCRUZ Paraná, Curitiba, Paraná, Brazil
| | - Thiago Neves Machado
- Laboratório FotoNanoBio, Universidade Tecnológica Federal do Paraná, Curitiba, Paraná, Brazil
| | - Arandi Ginane Bezerra
- Laboratório FotoNanoBio, Universidade Tecnológica Federal do Paraná, Curitiba, Paraná, Brazil
| | | | - Alessandra Melo de Aguiar
- Laboratório de Biologia Básica de Células-Tronco, Instituto Carlos Chagas, FIOCRUZ Paraná, Curitiba, Paraná, Brazil; Rede de Plataformas Tecnológicas FIOCRUZ - Bioensaios com Métodos Alternativos em Citotoxicidade, Instituto Carlos Chagas, FIOCRUZ Paraná, Curitiba, Paraná, Brazil.
| |
Collapse
|
15
|
Ferrarini MG, Nisimura LM, Girard RMBM, Alencar MB, Fragoso MSI, Araújo-Silva CA, Veiga ADA, Abud APR, Nardelli SC, Vommaro RC, Silber AM, France-Sagot M, Ávila AR. Dichloroacetate and Pyruvate Metabolism: Pyruvate Dehydrogenase Kinases as Targets Worth Investigating for Effective Therapy of Toxoplasmosis. mSphere 2021; 6:e01002-20. [PMID: 33408226 PMCID: PMC7845590 DOI: 10.1128/msphere.01002-20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 12/09/2020] [Indexed: 12/31/2022] Open
Abstract
Toxoplasmosis, a protozoan infection caused by Toxoplasma gondii, is estimated to affect around 2.5 billion people worldwide. Nevertheless, the side effects of drugs combined with the long period of therapy usually result in discontinuation of the treatment. New therapies should be developed by exploring peculiarities of the parasite's metabolic pathways, similarly to what has been well described in cancer cell metabolism. An example is the switch in the metabolism of cancer that blocks the conversion of pyruvate into acetyl coenzyme A in mitochondria. In this context, dichloroacetate (DCA) is an anticancer drug that reverts the tumor proliferation by inhibiting the enzymes responsible for this switch: the pyruvate dehydrogenase kinases (PDKs). DCA has also been used in the treatment of certain symptoms of malaria; however, there is no evidence of how this drug affects apicomplexan species. In this paper, we studied the metabolism of T. gondii and demonstrate that DCA also inhibits T. gondii's in vitro infection with no toxic effects on host cells. DCA caused an increase in the activity of pyruvate dehydrogenase followed by an unbalanced mitochondrial activity. We also observed morphological alterations frequently in mitochondria and in a few apicoplasts, essential organelles for parasite survival. To date, the kinases that potentially regulate the activity of pyruvate metabolism in both organelles have never been described. Here, we confirmed the presence in the genome of two putative kinases (T. gondii PDK [TgPDK] and T. gondii branched-chain α-keto acid dehydrogenase kinase [TgBCKDK]), verified their cellular localization in the mitochondrion, and provided in silico data suggesting that they are potential targets of DCA.IMPORTANCE Currently, the drugs used for toxoplasmosis have severe toxicity to human cells, and the treatment still lacks effective and safer alternatives. The search for novel drug targets is timely. We report here that the treatment of T. gondii with an anticancer drug, dichloroacetate (DCA), was effective in decreasing in vitro infection without toxicity to human cells. It is known that PDK is the main target of DCA in mammals, and this inactivation increases the conversion of pyruvate into acetyl coenzyme A and reverts the proliferation of tumor cells. Moreover, we verified the mitochondrial localization of two kinases that possibly regulate the activity of pyruvate metabolism in T. gondii, which has never been studied. DCA increased pyruvate dehydrogenase (PDH) activity in T. gondii, followed by an unbalanced mitochondrial activity, in a manner similar to what was previously observed in cancer cells. Thus, we propose the conserved kinases as potential regulators of pyruvate metabolism and interesting targets for new therapies.
Collapse
Affiliation(s)
- Mariana Galvão Ferrarini
- Laboratoire de Biométrie et Biologie Évolutive, UMR 5558, CNRS, Université de Lyon 1, Villeurbanne, France
| | - Lindice Mitie Nisimura
- Laboratório de Pesquisa em Apicomplexa, Instituto Carlos Chagas, Fiocruz, Paraná, Brazil
| | - Richard Marcel Bruno Moreira Girard
- Laboratory of Biochemistry of Tryps, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Mayke Bezerra Alencar
- Laboratory of Biochemistry of Tryps, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | | | - Carlla Assis Araújo-Silva
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Biologia Estrutural e Bioimagens, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alan de Almeida Veiga
- Laboratório de Pesquisa em Apicomplexa, Instituto Carlos Chagas, Fiocruz, Paraná, Brazil
| | | | | | - Rossiane C Vommaro
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Biologia Estrutural e Bioimagens, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ariel Mariano Silber
- Laboratory of Biochemistry of Tryps, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Marie France-Sagot
- Laboratoire de Biométrie et Biologie Évolutive, UMR 5558, CNRS, Université de Lyon 1, Villeurbanne, France
- INRIA Grenoble Rhône-Alpes, Montbonnot-Saint-Martin, France
| | - Andréa Rodrigues Ávila
- Laboratório de Pesquisa em Apicomplexa, Instituto Carlos Chagas, Fiocruz, Paraná, Brazil
| |
Collapse
|
16
|
Robert AW, Stimamiglio MA. The secretome from embryonic stem cell cardiomyogenesis: Same signals, different cellular feedbacks. J Cell Physiol 2020; 236:971-980. [PMID: 32592189 DOI: 10.1002/jcp.29907] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/26/2020] [Accepted: 06/16/2020] [Indexed: 12/15/2022]
Abstract
Ischemic heart diseases are a global health problem that requires the search for alternative therapies to the current treatments. Thus, an understanding of how cardiomyogenic signals can affect cellular behavior would allow us to create strategies to improve the cell recovery in damaged tissues. In this study, we aimed to evaluate the effects of the conditioned medium (CM), collected at different time points during in vitro cardiomyogenesis of human embryonic stem cells (hESCs), to direct cell behavior. We assayed different cell types to demonstrate noncytotoxic effects from the collected CM and that the CM obtained at initial time points of cardiomyogenic differentiation could promote the cell proliferation. Otherwise, the secretome derived from cardiac committed cells during cardiomyogenesis was unable to improve angiogenesis or migration in endothelial cells, and ineffective to stimulate the differentiation of cardioblasts or increase the differentiation efficiency of hESC. Therefore, we demonstrated that the effectiveness of the CM response varies depending on the cell type and the differentiation step of hESC-derived cardiomyocytes.
Collapse
Affiliation(s)
- Anny W Robert
- Laboratório de Biologia Básica de Células-Tronco, Instituto Carlos Chagas, Fiocruz-Paraná, Curitiba, Paraná, Brazil
| | - Marco A Stimamiglio
- Laboratório de Biologia Básica de Células-Tronco, Instituto Carlos Chagas, Fiocruz-Paraná, Curitiba, Paraná, Brazil
| |
Collapse
|
17
|
Dose-dependent cell necrosis induced by silica nanoparticles. Toxicol In Vitro 2020; 63:104723. [DOI: 10.1016/j.tiv.2019.104723] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 10/30/2019] [Accepted: 11/14/2019] [Indexed: 11/20/2022]
|
18
|
Abud APR, Kuligovski C, Corrêa NCR, de Moraes ECP, Caruso RRB, Schuck DC, Brohem CA, Dallagiovanna B, de Aguiar AM. The inhibition of adipogenesis via an in vitro assay can reduce animal use by more precisely estimating the starting dose for the acute toxic class method. Toxicol Lett 2019; 311:80-90. [PMID: 31029752 DOI: 10.1016/j.toxlet.2019.04.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 03/01/2019] [Accepted: 04/08/2019] [Indexed: 02/07/2023]
Abstract
In the present work, we established an adipogenesis inhibition assay as an adequate and sensitive in vitro model for reducing animal use by estimating the starting dose for the acute toxic class (ATC) method. First, human adipose-derived stem cells (ADSCs) underwent adipogenic differentiation induction for 14 days. Then, by high-content imaging analysis, we determined the percentage and area of cell differentiation that we considered suitable for negative and positive internal control according to the quality control criteria strictly standardized mean difference (SSMD) and robust SSMD. Moreover, we established sodium dodecyl sulfate (SDS) as an external positive control in this assay. To measure reduction in animal use to estimate the starting dose for the ATC method, we evaluated 10 chemicals representing Globally Harmonized System of Classification and Labeling of Chemicals (GHS) toxicity categories 1-5 and unclassified toxicity and determined the dose-response curves for percentage and area of cell differentiation by using the Hill function with an R2 ≥ 0.85. The resulting IC50 values were used for LD50 prediction and for estimating the starting dose for the ATC method. Our results indicated that use of the inhibition of adipogenesis assay to estimate the starting dose for the ATC method would decrease animal use for 7 out of 10 tested substances, possibly all substances if we consider the more toxic test substances in GHS categories 1, 2, and 3. We can conclude that the present assay is a suitable alternative to reduce animal testing in the first steps of predicting highly toxic substances. Moreover, this method also presents internal and external controls as differentials, which guarantee the quality of the assay as well as the results. These features are important for suggesting a methodology for regulatory purposes.
Collapse
Affiliation(s)
- Ana Paula Ressett Abud
- Laboratório de Biologia Básica de Células-Tronco, Instituto Carlos Chagas, Fiocruz, Curitiba, PR, Brazil
| | - Crisciele Kuligovski
- Laboratório de Biologia Básica de Células-Tronco, Instituto Carlos Chagas, Fiocruz, Curitiba, PR, Brazil
| | | | - Elizabeth Cunha Penna de Moraes
- Laboratório de Biologia Básica de Células-Tronco, Instituto Carlos Chagas, Fiocruz, Curitiba, PR, Brazil; Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Rodrigo Rêgo Barros Caruso
- Laboratório de Biologia Básica de Células-Tronco, Instituto Carlos Chagas, Fiocruz, Curitiba, PR, Brazil; Instituto Carlos Chagas, Fiocruz, Curitiba, PR, Brazil
| | | | - Carla Abdo Brohem
- Núcleo de Avaliação de Segurança (Grupo Boticário), Curitiba, PR, Brazil
| | - Bruno Dallagiovanna
- Laboratório de Biologia Básica de Células-Tronco, Instituto Carlos Chagas, Fiocruz, Curitiba, PR, Brazil.
| | - Alessandra Melo de Aguiar
- Laboratório de Biologia Básica de Células-Tronco, Instituto Carlos Chagas, Fiocruz, Curitiba, PR, Brazil.
| |
Collapse
|
19
|
Kim TW, Che JH, Yun JW. Use of stem cells as alternative methods to animal experimentation in predictive toxicology. Regul Toxicol Pharmacol 2019; 105:15-29. [DOI: 10.1016/j.yrtph.2019.03.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 03/23/2019] [Accepted: 03/25/2019] [Indexed: 12/16/2022]
|
20
|
Amara R, Awad H, Chaker D, Bentabed‐Ababsa G, Lassagne F, Erb W, Chevallier F, Roisnel T, Dorcet V, Fajloun Z, Vidal J, Mongin F. Conversion of Isatins to Tryptanthrins, Heterocycles Endowed with a Myriad of Bioactivities. European J Org Chem 2019. [DOI: 10.1002/ejoc.201900352] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Rim Amara
- Univ Rennes, CNRS ISCR (Institut des Sciences Chimiques de Rennes) ‐ UMR 6226 35000 Rennes France
- Laboratoire de Synthèse Organique Appliquée Faculté des Sciences Exactes et Appliquées Université Oran1 Ahmed Ben Bella BP 1524 El M'Naouer 31000 Oran Algeria
| | - Haçan Awad
- Faculty of Sciences 3 Lebanese University Campus El‐Kobbeh Tripoli Lebanon
| | - Diana Chaker
- Laboratory of Applied Biotechnology Azm Center for Research in Biotechnology and its Applications, EDST Lebanese University 1300 Tripoli Lebanon
| | - Ghenia Bentabed‐Ababsa
- Laboratoire de Synthèse Organique Appliquée Faculté des Sciences Exactes et Appliquées Université Oran1 Ahmed Ben Bella BP 1524 El M'Naouer 31000 Oran Algeria
| | - Frédéric Lassagne
- Univ Rennes, CNRS ISCR (Institut des Sciences Chimiques de Rennes) ‐ UMR 6226 35000 Rennes France
| | - William Erb
- Univ Rennes, CNRS ISCR (Institut des Sciences Chimiques de Rennes) ‐ UMR 6226 35000 Rennes France
| | - Floris Chevallier
- Univ Rennes, CNRS ISCR (Institut des Sciences Chimiques de Rennes) ‐ UMR 6226 35000 Rennes France
| | - Thierry Roisnel
- Univ Rennes, CNRS ISCR (Institut des Sciences Chimiques de Rennes) ‐ UMR 6226 35000 Rennes France
| | - Vincent Dorcet
- Univ Rennes, CNRS ISCR (Institut des Sciences Chimiques de Rennes) ‐ UMR 6226 35000 Rennes France
| | - Ziad Fajloun
- Faculty of Sciences 3 Lebanese University Campus El‐Kobbeh Tripoli Lebanon
- Laboratory of Applied Biotechnology Azm Center for Research in Biotechnology and its Applications, EDST Lebanese University 1300 Tripoli Lebanon
| | - Joëlle Vidal
- Univ Rennes, CNRS ISCR (Institut des Sciences Chimiques de Rennes) ‐ UMR 6226 35000 Rennes France
| | - Florence Mongin
- Univ Rennes, CNRS ISCR (Institut des Sciences Chimiques de Rennes) ‐ UMR 6226 35000 Rennes France
| |
Collapse
|
21
|
de Castro E, Reus TL, de Aguiar AM, Ávila AR, de Arruda Campos Brasil de Souza T. Procaspase-activating compound-1 induces apoptosis in Trypanosoma cruzi. Apoptosis 2018; 22:1564-1577. [PMID: 29058102 DOI: 10.1007/s10495-017-1428-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Some therapeutics for parasitic, cardiac and neurological diseases activate apoptosis. Therefore, the study of apoptotic proteins in pathogenic organisms is relevant. However, the molecular mechanism of apoptosis in unicellular organisms remain elusive, despite morphological evidence of its occurrence. In Trypanosoma cruzi, the causative agent of Chagas disease, metacaspase 3 (TcMCA3), seems to have a key role in parasite apoptosis. Accordingly, this work provides data concerning TcMCA3 regulation through its interaction with procaspase-activating compound 1 (PAC-1), a procaspase 3 activator. Indeed, PAC-1 reduced T. cruzi epimastigote viability with an IC50 of 14.12 µM and induced loss of mitochondrial potential and exposure of phosphatidylserine, features of the apoptotic process. Notwithstanding, those PAC-1-inducible effects were not conserved in metacyclic trypomastigotes. Moreover, PAC-1 reduced the viability of mammalian cells with a greater IC50 (25.70 µM) compared to T. cruzi epimastigotes, indicating distinct modes of binding between caspases and metacaspases. To shed light on the selectivity of metacaspases and caspases, we determined the structural features related to the PAC-1 binding sites in both types of proteins. These data are important for improving the understanding of the apoptosis pathway in T. cruzi so that TcMCA3 could be better targeted with future pharmaceuticals.
Collapse
Affiliation(s)
- Emanuella de Castro
- Laboratório de Proteômica e Engenharia de Proteínas, Instituto Carlos Chagas, Fiocruz, Curitiba, Brazil
| | - Thamile Luciane Reus
- Laboratório de Biologia Básica de Células Tronco, Instituto Carlos Chagas, Fiocruz, Curitiba, PR, Brazil
| | - Alessandra Melo de Aguiar
- Laboratório de Biologia Básica de Células Tronco, Instituto Carlos Chagas, Fiocruz, Curitiba, PR, Brazil
| | - Andrea Rodrigues Ávila
- Laboratório de Regulação da Expressão Gênica, Instituto Carlos Chagas, Fiocruz, Curitiba, PR, Brazil
| | | |
Collapse
|
22
|
Reus TL, Machado TN, Bezerra AG, Marcon BH, Paschoal ACC, Kuligovski C, de Aguiar AM, Dallagiovanna B. Dose-dependent cytotoxicity of bismuth nanoparticles produced by LASiS in a reference mammalian cell line BALB/c 3T3. Toxicol In Vitro 2018; 53:99-106. [PMID: 30030050 DOI: 10.1016/j.tiv.2018.07.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 06/11/2018] [Accepted: 07/06/2018] [Indexed: 12/24/2022]
Abstract
Nanoparticles (NPs) have emerged as new potential tools for many applications in previous years. Among all types of NPs, bismuth NPs (BiNPs) have a very low cost and potential for many applications, ranging from medicine to industry. Although the toxic effects of bismuth have been studied, little is known about its toxicity at the nanoscale level. Therefore, in this study, we aimed to investigate the cytotoxic effects of BiNPs produced by laser ablation synthesis in solution (LASiS) in a reference mammalian cell line to evaluate their cytotoxicity (BALB/c 3 T3 cells). We also stabilized BiNPs in two different solutions: culture medium supplemented with fetal bovine serum (FBS) and bovine serum albumin (BSA). The cytotoxicity of BiNPs in culture medium (IC50:28.51 ± 9.96 μg/ml) and in BSA (IC50:25.54 ± 8.37 μg/ml) was assessed, and they were not significantly different. Second, the LD50 was predicted, and BiNPs were estimated as GHS class 4. We also found that cell death occurs due to apoptosis. By evaluating the interaction between BiNPs and cells at ultrastructural level, we suggest that cell death occurs once BiNPs are internalized. Additionally, we suggest that BiNPs cause cell damage because myelin figures were found inside cells that had internalized BiNPs. To date, this is the first study to assess the cytotoxicity of BiNPs produced by LASiS and to predict the possible LD50 and GHS class of BiNPs.
Collapse
Affiliation(s)
- Thamile Luciane Reus
- Laboratório de Biologia Básica de Células-tronco, Instituto Carlos Chagas, FIOCRUZ Paraná, Rua Prof. Algacyr Munhoz Mader, 3775 CIC, 81350-010 Curitiba, PR, Brazil
| | - Thiago Neves Machado
- Laboratório FOTONANOBIO, Universidade Tecnológica Federal do Paraná, Avenida 7 de Setembro 3165, 80230-901 Curitiba, PR, Brazil
| | - Arandi Ginane Bezerra
- Laboratório FOTONANOBIO, Universidade Tecnológica Federal do Paraná, Avenida 7 de Setembro 3165, 80230-901 Curitiba, PR, Brazil
| | - Bruna Hilzendeger Marcon
- Laboratório de Biologia Básica de Células-tronco, Instituto Carlos Chagas, FIOCRUZ Paraná, Rua Prof. Algacyr Munhoz Mader, 3775 CIC, 81350-010 Curitiba, PR, Brazil
| | - Ariane Caroline Campos Paschoal
- Laboratório de Biologia Básica de Células-tronco, Instituto Carlos Chagas, FIOCRUZ Paraná, Rua Prof. Algacyr Munhoz Mader, 3775 CIC, 81350-010 Curitiba, PR, Brazil
| | - Crisciele Kuligovski
- Laboratório de Biologia Básica de Células-tronco, Instituto Carlos Chagas, FIOCRUZ Paraná, Rua Prof. Algacyr Munhoz Mader, 3775 CIC, 81350-010 Curitiba, PR, Brazil
| | - Alessandra Melo de Aguiar
- Laboratório de Biologia Básica de Células-tronco, Instituto Carlos Chagas, FIOCRUZ Paraná, Rua Prof. Algacyr Munhoz Mader, 3775 CIC, 81350-010 Curitiba, PR, Brazil.
| | - Bruno Dallagiovanna
- Laboratório de Biologia Básica de Células-tronco, Instituto Carlos Chagas, FIOCRUZ Paraná, Rua Prof. Algacyr Munhoz Mader, 3775 CIC, 81350-010 Curitiba, PR, Brazil.
| |
Collapse
|
23
|
Corrêa NCR, Kuligovski C, Paschoal ACC, Abud APR, Rebelatto CLK, Leite LMB, Senegaglia AC, Dallagiovanna B, Aguiar AMD. Human adipose-derived stem cells (ADSC) and human periodontal ligament stem cells (PDLSC) as cellular substrates of a toxicity prediction assay. Regul Toxicol Pharmacol 2018; 92:75-82. [DOI: 10.1016/j.yrtph.2017.11.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 11/07/2017] [Accepted: 11/08/2017] [Indexed: 01/08/2023]
|
24
|
Huang T, Yan L, Zheng S, Wang Y, Wang X, Fan L, Li C, Zhao Y, Martyniuk CJ. Discriminating modes of toxic action in mice using toxicity in BALB/c mouse fibroblast (3T3) cells. CHEMOSPHERE 2017; 188:73-80. [PMID: 28869848 DOI: 10.1016/j.chemosphere.2017.08.135] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 08/22/2017] [Accepted: 08/26/2017] [Indexed: 06/07/2023]
Abstract
The objective of this study was to determine whether toxicity in mouse fibroblast cells (3T3 cells) could predict toxicity in mice. Synthesized data on toxicity was subjected to regression analysis and it was observed that relationship of toxicities between mice and 3T3 cells was not strong (R2 = 0.41). Inclusion of molecular descriptors (e.g. ionization, pKa) improved the regression to R2 = 0.56, indicating that this relationship is influenced by kinetic processes of chemicals or specific toxic mechanisms associated to the compounds. However, to determine if we were able to discriminate modes of action (MOAs) in mice using the toxicities generated from 3T3 cells, compounds were first classified into "baseline" and "reactive" guided by the toxic ratio (TR) for each compound in mice. Sequence, binomial and recursive partitioning analyses provided strong predictions of MOAs in mice based upon toxicities in 3T3 cells. The correct classification of MOAs based on these methods was 86%. Nearly all the baseline compounds predicted from toxicities in 3T3 cells were identified as baseline compounds from the TR in mice. The incorrect assignment of MOAs for some compounds is hypothesized to be due to experimental uncertainty that exists in toxicity assays for both mice and 3T3 cells. Conversely, lack of assignment can also arise because some reactive compounds have MOAs that are different in mice compared to 3T3 cells. The methods developed here are novel and contribute to efforts to reduce animal numbers in toxicity tests that are used to evaluate risks associated with organic pollutants in the environment.
Collapse
Affiliation(s)
- Tao Huang
- State Environmental Protection Key Laboratory of Wetland Ecology and Vegetation Restoration, School of Environment, Northeast Normal University, Changchun, Jilin 130117, PR China
| | - Lichen Yan
- State Environmental Protection Key Laboratory of Wetland Ecology and Vegetation Restoration, School of Environment, Northeast Normal University, Changchun, Jilin 130117, PR China
| | - Shanshan Zheng
- State Environmental Protection Key Laboratory of Wetland Ecology and Vegetation Restoration, School of Environment, Northeast Normal University, Changchun, Jilin 130117, PR China
| | - Yue Wang
- State Environmental Protection Key Laboratory of Wetland Ecology and Vegetation Restoration, School of Environment, Northeast Normal University, Changchun, Jilin 130117, PR China
| | - Xiaohong Wang
- State Environmental Protection Key Laboratory of Wetland Ecology and Vegetation Restoration, School of Environment, Northeast Normal University, Changchun, Jilin 130117, PR China; Center for Environmental and Human Toxicology, Department of Physiological Sciences, College of Veterinary Medicine, UF Genetics Institute, Interdisciplinary Program in Biomedical Sciences Neuroscience, University of Florida, Gainesville, FL 32611, USA
| | - Lingyun Fan
- State Environmental Protection Key Laboratory of Wetland Ecology and Vegetation Restoration, School of Environment, Northeast Normal University, Changchun, Jilin 130117, PR China
| | - Chao Li
- State Environmental Protection Key Laboratory of Wetland Ecology and Vegetation Restoration, School of Environment, Northeast Normal University, Changchun, Jilin 130117, PR China
| | - Yuanhui Zhao
- State Environmental Protection Key Laboratory of Wetland Ecology and Vegetation Restoration, School of Environment, Northeast Normal University, Changchun, Jilin 130117, PR China.
| | - Christopher J Martyniuk
- Center for Environmental and Human Toxicology, Department of Physiological Sciences, College of Veterinary Medicine, UF Genetics Institute, Interdisciplinary Program in Biomedical Sciences Neuroscience, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
25
|
Establishment and evaluation of immortalized human epidermal keratinocytes for an alternative skin irritation test. J Pharmacol Toxicol Methods 2017; 88:130-139. [PMID: 28827132 DOI: 10.1016/j.vascn.2017.08.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 06/24/2017] [Accepted: 08/18/2017] [Indexed: 12/11/2022]
Abstract
Human skin is located at the outermost part of the body, and various cosmetics and chemicals that may come in contact with human skin need to be evaluated for their potential to cause irritation. Until recently, the Draize test was considered the standard method for skin irritation; however, this technique has disadvantages such as the need to sacrifice many rabbits and subjective scoring. Thus, to contribute to the development of an animal-free alternative skin irritation test, we investigated the cytotoxicity and inflammatory response to standard skin irritants in SV40 large T antigen-transformed human epidermal keratinocyte 2 cells (SV-HEK2 cells). In this study, we established an SV-HEK2 cell line immortalized by SV40 large T antigen (SV40 T) and characterized the inherent morphological and cytological properties. We next used 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) or neutral red uptake (NRU) assays of cell viability to investigate the optimal experimental conditions for determining SV-HEK2 cell viability after exposure to sodium dodecyl sulfate at 6.25×10-3% to 1×10-1% as a standard skin irritant. We then examined the viability of SV-HEK2 cells in response to five skin irritants (benzalkonium chloride, isopropanol, sodium dodecyl sulfate, Triton X-100 and Tween20) at 5×10-3% to 1×10-1% by MTT or NRU assay. Finally, we estimated the level of cytokines secretion in response to stimulation by skin irritants in SV-HEK2 cells. The results revealed that SV-HEK2 cells responded well to skin irritants in a concentration-dependent manner and that there was good correlation between irritant concentration and cytotoxicity (or cytokine secretion) when cells were exposed to skin irritants for 10min at room temperature (RT) using an MTT assay. Overall, these findings suggest that SV-HEK2 cells could be a good alternative in vitro model for skin irritation tests.
Collapse
|
26
|
Cytotoxicity of GuttaFlow Bioseal, GuttaFlow2, MTA Fillapex, and AH Plus on Human Periodontal Ligament Stem Cells. J Endod 2017; 43:816-822. [DOI: 10.1016/j.joen.2017.01.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 12/30/2016] [Accepted: 01/02/2017] [Indexed: 11/17/2022]
|
27
|
Cui JW, Wang Q, Bi S, Zhang J, Zhu JL, Liu JG, Wu DM. Construction and application of a new cell electrochemical detecting system based on the hyposmotic principle. Analyst 2017; 142:591-595. [PMID: 28121318 DOI: 10.1039/c6an02114j] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A new cell electrochemical detecting system has been constructed based on the hyposmotic principle, in which the electrochemical signals have been strengthened by about 109.75% for the signal at about +0.70 V and 532.94% for the signal at about +1.03 V. The electrochemical detection limits of the cells have been improved by one order of magnitude. The individual concentrations of intracellular purines have been obtained.
Collapse
Affiliation(s)
- Ji-Wen Cui
- College of Pharmacy, Jiamusi University, Jiamusi, 154000, China.
| | | | | | | | | | | | | |
Collapse
|
28
|
Secretome from resident cardiac stromal cells stimulates proliferation, cardiomyogenesis and angiogenesis of progenitor cells. Int J Cardiol 2016; 221:396-403. [PMID: 27404713 DOI: 10.1016/j.ijcard.2016.06.199] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 06/09/2016] [Accepted: 06/25/2016] [Indexed: 12/20/2022]
Abstract
In the heart, tissue-derived signals play a central role on recruiting/activating stem cell sources to induce cardiac lineage specification for maintenance of tissue homeostasis and repair. Cardiac resident stromal cells (CRSCs) may play a pivotal role in cardiac repair throughout their secretome. Here, we performed the characterization of CRSCs and their secretome by analyzing the composition of their culture-derived extracellular matrix (ECM) and conditioned medium (CM) and by investigating their potential effect on adipose-derived stem cell (ADSC) and progenitor cell behavior. We confirmed that CRSCs are a heterogeneous cell population whose secretome is composed by proteins related to cellular growth, immune response and cardiovascular development and function. We also observed that CRSC secretome was unable to change the behavior of ADSCs, except for proliferation. Additionally, CM from CRSCs demonstrated the potential to drive proliferation and cardiac differentiation of H9c2 cells and also the ability to induce angiogenesis in vitro. Our data suggest that the CRSCs can be a source of important modulating signals for cardiac progenitor cell recruitment/activation.
Collapse
|
29
|
Zhang Y, Yin J, Ding H, Zhang C, Gao YS. Vitamin K2 Ameliorates Damage of Blood Vessels by Glucocorticoid: a Potential Mechanism for Its Protective Effects in Glucocorticoid-induced Osteonecrosis of the Femoral Head in a Rat Model. Int J Biol Sci 2016; 12:776-85. [PMID: 27313492 PMCID: PMC4910597 DOI: 10.7150/ijbs.15248] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 03/25/2016] [Indexed: 12/22/2022] Open
Abstract
Glucocorticoid has been reported to decrease blood vessel number and harm the blood supply in the femoral head, which is recognized to be an important mechanism of glucocorticoid-induced osteonecrosis of the femoral head (ONFH). To prevent glucocorticoid-induced ONFH, medication that promotes both bone formation and angiogenesis would be ideal. Vitamin K2 has been revealed to play an important role in bone metabolism; however, few studies have focused on the effect of Vitamin K2 on new vascular formation. Thus, this study aimed to investigate whether Vitamin K2 promoted new blood vessel formation in the presence of glucocorticoids, both in vitro and in vivo. The effect of Vitamin K2 on viability, migration, in vitro tube formation, and VEGF, vWF, CD31, KDR, Flt and PDGFB in EAhy926 incubated with or without dexamethasone were elucidated. VEGF, TGF-β and BMP-2, angiogenesis-related proteins secreted by osteoblasts, were also detected in the osteoblast-like cell line of MG63. In addition, blood vessels of the femoral head in rats administered with or without methylprednisolone and Vitamin K2 were evaluated using angiography and CD31 staining. In vitro studies showed that Vitamin K2 significantly protected endothelial cells from dexamethasone-induced apoptosis, promoted endothelial cell migration and in vitro tube formation. Angiogenesis-related proteins both in EAhy926 and MG63 were also upregulated by Vitamin K2 when cotreated with dexamethasone. In vivo studies showed enhanced blood vessel volume and CD31-positive staining cells in rats cotreated with VK2 and methylprednisolone compared to rats treated with methylprednisolone only. Collectively, Vitamin K2 has the ability to promote angiogenesis in vitro and to ameliorate vessels of the femoral head in glucocorticoid-treated rats in vivo, indicating that Vitamin K2 is a promising drug that may be used to prevent steroid-induced ONFH.
Collapse
Affiliation(s)
- Yuelei Zhang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Junhui Yin
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Hao Ding
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Changqing Zhang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - You-Shui Gao
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|