1
|
Suominen A, Suni A, Ruohonen S, Szabó Z, Pohjolainen L, Cai M, Savontaus E, Talman V, Kerkelä R, Rinne P. Melanocortin 1 Receptor Regulates Pathological and Physiological Cardiac Remodeling. J Am Heart Assoc 2025; 14:e037961. [PMID: 39921516 PMCID: PMC12074753 DOI: 10.1161/jaha.124.037961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 11/22/2024] [Indexed: 02/10/2025]
Abstract
BACKGROUND The melanocortin 1 receptor (MC1R) is abundantly expressed in the skin and leukocytes, where it regulates skin pigmentation and inflammatory responses. Recently, MC1R was also found in the heart, but its functional role has remained unknown. We aimed to investigate whether MC1R is involved in the regulation of pathological or physiological cardiac remodeling. METHODS AND RESULTS Recessive yellow mice, as a model of global MC1R deficiency, and cardiomyocyte-specific MC1R knockout mice were subjected to transverse aortic constriction or voluntary wheel running to induce pathological or physiological cardiac hypertrophy, respectively. Mice were phenotyped for cardiac structure and function by echocardiography, histology, and quantitative PCR analysis. H9c2 cells and neonatal mouse ventricular cardiac myocytes were used as in vitro models to investigate the effects of pharmacological MC1R activation on hypertrophy-related responses. We found that the expression of MC1R progressively declines in the failing mouse heart. MC1R recessive yellow mice showed blunted hypertrophic response to transverse aortic constriction-induced pressure overload and exercise training. This phenotype was recapitulated in MC1R knockout mice, demonstrating that MC1R deficiency specifically in cardiomyocytes is responsible for the antihypertrophic effect. However, MC1R knockout mice subjected to pressure overload showed left ventricular dilatation that was associated with reduced ejection fraction and changes in left ventricular diastolic function. At the molecular level, the mRNA expression of myosin heavy chain β was upregulated in the hearts of MC1R knockout mice. In contrast, selective activation of MC1R promoted hypertrophic responses in cultured cardiomyocytes. CONCLUSIONS Cardiomyocyte-specific MC1R deficiency attenuates physiological and pathological cardiac hypertrophy in mice, while pharmacological activation of MC1R promotes cardiomyocyte hypertrophy.
Collapse
MESH Headings
- Animals
- Ventricular Remodeling
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Mice, Knockout
- Receptor, Melanocortin, Type 1/genetics
- Receptor, Melanocortin, Type 1/metabolism
- Receptor, Melanocortin, Type 1/deficiency
- Disease Models, Animal
- Mice
- Cardiomegaly/metabolism
- Cardiomegaly/physiopathology
- Cardiomegaly/genetics
- Cardiomegaly/pathology
- Male
- Hypertrophy, Left Ventricular/metabolism
- Hypertrophy, Left Ventricular/physiopathology
- Hypertrophy, Left Ventricular/genetics
- Hypertrophy, Left Ventricular/pathology
- Heart Failure/metabolism
- Heart Failure/physiopathology
- Heart Failure/genetics
- Heart Failure/pathology
- Ventricular Function, Left
- Mice, Inbred C57BL
- Rats
- Phenotype
Collapse
Affiliation(s)
- Anni Suominen
- Research Centre for Integrative Physiology & Pharmacology, Institute of BiomedicineUniversity of TurkuFinland
- Drug Research Doctoral Programme (DRDP)University of TurkuFinland
| | - Aino Suni
- Research Centre for Integrative Physiology & Pharmacology, Institute of BiomedicineUniversity of TurkuFinland
| | - Saku Ruohonen
- Research Centre for Integrative Physiology & Pharmacology, Institute of BiomedicineUniversity of TurkuFinland
| | - Zoltán Szabó
- Research Unit of Biomedicine and Internal Medicine, Department of Pharmacology and ToxicologyUniversity of OuluFinland
| | - Lotta Pohjolainen
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of PharmacyUniversity of HelsinkiFinland
| | - Minying Cai
- Department of Chemistry and BiochemistryUniversity of ArizonaTucsonAZUSA
| | - Eriika Savontaus
- Research Centre for Integrative Physiology & Pharmacology, Institute of BiomedicineUniversity of TurkuFinland
- Turku Center for Disease ModelingUniversity of TurkuFinland
- Unit of Clinical PharmacologyTurku University HospitalTurkuFinland
| | - Virpi Talman
- Research Centre for Integrative Physiology & Pharmacology, Institute of BiomedicineUniversity of TurkuFinland
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of PharmacyUniversity of HelsinkiFinland
| | - Risto Kerkelä
- Research Unit of Biomedicine and Internal Medicine, Department of Pharmacology and ToxicologyUniversity of OuluFinland
- Medical Research Center OuluOulu University Hospital and University of OuluFinland
| | - Petteri Rinne
- Research Centre for Integrative Physiology & Pharmacology, Institute of BiomedicineUniversity of TurkuFinland
- Turku Center for Disease ModelingUniversity of TurkuFinland
| |
Collapse
|
2
|
Kim SR, Min EK, Lee CM, Lee JW, Park CH, Jung Y, Oh BC, Lee HY. Development of Advanced Oral-on-a-Chip: Replicating the Intricate Human Oral Microenvironment. Int J Biol Sci 2024; 20:5888-5909. [PMID: 39664582 PMCID: PMC11628345 DOI: 10.7150/ijbs.104351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 10/22/2024] [Indexed: 12/13/2024] Open
Abstract
The interactions between various cellular populations in the oral cavity, including gingival keratinocytes, tonsil-resident stem cells, periodontal ligament fibroblasts, and vascular endothelial cells, are crucial for maintaining oral health. These interactions regulate essential functions like tooth support and pathogen defense. However, conventional 2D and 3D in vitro models often fail to capture the complexity of these interactions and the multicellular architecture of the oral environment. To address this limitation, we developed an advanced 3D oral-on-a-chip system that mimics the dynamic microenvironment of oral tissues. This system incorporates multiple oral cells into a 3D structure made from natural polymers such as collagen and hyaluronic acid, crosslinked by blood-coagulating factors. Our study revealed that tonsil-resident stem cells are more sensitive to toxic exposure compared to differentiated cells like fibroblasts and endothelial cells. SERPINB2 was identified as a key biomarker of oral toxicity, with significant upregulation observed in tonsil-resident stem cells after exposure to toxins. Based on this, we developed a fluorescence-linked toxicity detection system using SERPINB2, enabling sensitive and quantitative assessments of oral toxicity. This integrated system provides a valuable tool for evaluating the oral toxicity of drug candidates.
Collapse
Affiliation(s)
- Soo-Rim Kim
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, Republic of Korea
- Department of Molecular Medicine, School of Medicine, Gachon University, Incheon 406-840, Republic of Korea
| | - Eun-Kyung Min
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, Republic of Korea
- Department of Molecular Medicine, School of Medicine, Gachon University, Incheon 406-840, Republic of Korea
| | - Choon-Mi Lee
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, Republic of Korea
- Department of Molecular Medicine, School of Medicine, Gachon University, Incheon 406-840, Republic of Korea
| | - Jin Woo Lee
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, Republic of Korea
- Department of Molecular Medicine, School of Medicine, Gachon University, Incheon 406-840, Republic of Korea
| | - Chan Hum Park
- Department of Otolaryngology-Head and Neck Surgery, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Chuncheon, Republic of Korea
| | - YunJae Jung
- Department of Microbiology, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| | - Byung-Chul Oh
- Department of Physiology, Lee Gil Ya Cancer and Diabetes Institute, Gachon University College of Medicine, Incheon, 21999, Republic of Korea
| | - Hwa-Yong Lee
- Division of Science Education, Kangwon National University, 24341, Republic of Korea
| |
Collapse
|
3
|
Schaefer S, Melodia D, Corrigan N, Lenardon MD, Boyer C. Effect of Star Topology Versus Linear Polymers on Antifungal Activity and Mammalian Cell Toxicity. Macromol Biosci 2024; 24:e2300452. [PMID: 38009827 DOI: 10.1002/mabi.202300452] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/05/2023] [Indexed: 11/29/2023]
Abstract
The global increase in invasive fungal infections and the emergence of drug-resistant strains demand the urgent development of novel antifungal drugs. In this context, synthetic polymers with diverse compositions, mimicking natural antimicrobial peptides, have shown promising potential for combating fungal infections. This study investigates how altering polymer end-groups and topology from linear to branched star-like structures affects their efficacy against Candida spp., including clinical isolates. Additionally, the polymers' biocompatibility is accessed with murine embryonic fibroblasts and red blood cells in vitro. Notably, a low-molecular weight star polymer outperforms both its linear polymeric counterparts and amphotericin B (AmpB) in terms of an improved therapeutic index and reduced haemolytic activity, despite a higher minimum inhibitory concentration against Candida albicans (C. albicans) SC5314 (16-32 µg mL-1 vs 1 µg mL-1 for AmpB). These findings demonstrate the potential of synthetic polymers with diverse topologies as promising candidates for antifungal applications.
Collapse
Affiliation(s)
- Sebastian Schaefer
- School of Chemical Engineering, University of New South Wales (UNSW), Sydney, New South Wales, 2052, Australia
- Australian Centre for NanoMedicine, UNSW, Sydney, New South Wales, 2052, Australia
- School of Biotechnology and Biomolecular Sciences, UNSW, Sydney, New South Wales, 2052, Australia
| | - Daniele Melodia
- School of Chemical Engineering, University of New South Wales (UNSW), Sydney, New South Wales, 2052, Australia
- Australian Centre for NanoMedicine, UNSW, Sydney, New South Wales, 2052, Australia
| | - Nathaniel Corrigan
- School of Chemical Engineering, University of New South Wales (UNSW), Sydney, New South Wales, 2052, Australia
- Australian Centre for NanoMedicine, UNSW, Sydney, New South Wales, 2052, Australia
| | - Megan Denise Lenardon
- School of Biotechnology and Biomolecular Sciences, UNSW, Sydney, New South Wales, 2052, Australia
| | - Cyrille Boyer
- School of Chemical Engineering, University of New South Wales (UNSW), Sydney, New South Wales, 2052, Australia
- Australian Centre for NanoMedicine, UNSW, Sydney, New South Wales, 2052, Australia
| |
Collapse
|
4
|
Jumppanen M, Kinnunen SM, Zore M, Välimäki MJ, Talman V, Gennäs GBA, Ruskoaho HJ, Yli-Kauhaluoma J. Affinity chromatography reveals direct binding of the GATA4-NKX2-5 interaction inhibitor (3i-1000) with GATA4. Sci Rep 2024; 14:8938. [PMID: 38637629 PMCID: PMC11026519 DOI: 10.1038/s41598-024-59418-4] [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: 10/17/2023] [Accepted: 04/10/2024] [Indexed: 04/20/2024] Open
Abstract
Heart failure is a serious medical condition with a poor prognosis. Current treatments can only help manage the symptoms and slow the progression of heart failure. However, there is currently no cure to prevent and reverse cardiac remodeling. Transcription factors are in a central role in various cellular processes, and in the heart, GATA4 and NKX2-5 transcription factors mediate hypertrophic responses and remodeling. We have identified compounds that modulate the synergistic interaction of GATA4 and NKX2-5 and shown that the most promising compound (1, 3i-1000) is cardioprotective in vitro and in vivo. However, direct evidence of its binding site and mechanism of action has not been available. Due to the disordered nature of transcription factors, classical target engagement approaches cannot be utilized. Here, we synthesized a small-molecule ligand-binding pulldown probe of compound 1 to utilize affinity chromatography alongside CETSA, AlphaScreen, and molecular modeling to study ligand binding. These results provide the first evidence of direct physical binding of compound 1 selectively to GATA4. While developing drugs that target transcription factors presents challenges, advances in technologies and knowledge of intrinsically disordered proteins enable the identification of small molecules that can selectively target transcription factors.
Collapse
Affiliation(s)
- Mikael Jumppanen
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, (P.O. Box 56), FI-00014, Helsinki, Finland
| | - Sini M Kinnunen
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, (P.O. Box 56), FI-00014, Helsinki, Finland
| | - Matej Zore
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, (P.O. Box 56), FI-00014, Helsinki, Finland
| | - Mika J Välimäki
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, (P.O. Box 56), FI-00014, Helsinki, Finland
| | - Virpi Talman
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, (P.O. Box 56), FI-00014, Helsinki, Finland
| | - Gustav Boije Af Gennäs
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, (P.O. Box 56), FI-00014, Helsinki, Finland
| | - Heikki J Ruskoaho
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, (P.O. Box 56), FI-00014, Helsinki, Finland
| | - Jari Yli-Kauhaluoma
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, (P.O. Box 56), FI-00014, Helsinki, Finland.
| |
Collapse
|
5
|
Suominen A, Saldo Rubio G, Ruohonen S, Szabó Z, Pohjolainen L, Ghimire B, Ruohonen ST, Saukkonen K, Ijas J, Skarp S, Kaikkonen L, Cai M, Wardlaw SL, Ruskoaho H, Talman V, Savontaus E, Kerkelä R, Rinne P. α-Melanocyte-stimulating hormone alleviates pathological cardiac remodeling via melanocortin 5 receptor. EMBO Rep 2024; 25:1987-2014. [PMID: 38454158 PMCID: PMC11014855 DOI: 10.1038/s44319-024-00109-6] [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: 01/19/2024] [Revised: 01/23/2024] [Accepted: 02/16/2024] [Indexed: 03/09/2024] Open
Abstract
α-Melanocyte-stimulating hormone (α-MSH) regulates diverse physiological functions by activating melanocortin receptors (MC-R). However, the role of α-MSH and its possible target receptors in the heart remain completely unknown. Here we investigate whether α-MSH could be involved in pathological cardiac remodeling. We found that α-MSH was highly expressed in the mouse heart with reduced ventricular levels after transverse aortic constriction (TAC). Administration of a stable α-MSH analog protected mice against TAC-induced cardiac hypertrophy and systolic dysfunction. In vitro experiments revealed that MC5-R in cardiomyocytes mediates the anti-hypertrophic signaling of α-MSH. Silencing of MC5-R in cardiomyocytes induced hypertrophy and fibrosis markers in vitro and aggravated TAC-induced cardiac hypertrophy and fibrosis in vivo. Conversely, pharmacological activation of MC5-R improved systolic function and reduced cardiac fibrosis in TAC-operated mice. In conclusion, α-MSH is expressed in the heart and protects against pathological cardiac remodeling by activating MC5-R in cardiomyocytes. These results suggest that analogs of naturally occurring α-MSH, that have been recently approved for clinical use and have agonistic activity at MC5-R, may be of benefit in treating heart failure.
Collapse
Affiliation(s)
- Anni Suominen
- Research Centre for Integrative Physiology & Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
- Drug Research Doctoral Programme (DRDP), University of Turku, Turku, Finland
| | - Guillem Saldo Rubio
- Research Centre for Integrative Physiology & Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Saku Ruohonen
- Research Centre for Integrative Physiology & Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Zoltán Szabó
- Research Unit of Biomedicine and Internal Medicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Lotta Pohjolainen
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Bishwa Ghimire
- Institute for Molecular Medicine Finland (FIMM), HiLIFE Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Faculty of Medicine, University of Turku, Turku, Finland
| | - Suvi T Ruohonen
- Research Centre for Integrative Physiology & Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Karla Saukkonen
- Research Centre for Integrative Physiology & Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Jani Ijas
- Research Centre for Integrative Physiology & Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Sini Skarp
- Research Unit of Biomedicine and Internal Medicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Leena Kaikkonen
- Research Unit of Biomedicine and Internal Medicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Minying Cai
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ, USA
| | - Sharon L Wardlaw
- Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Heikki Ruskoaho
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Virpi Talman
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Eriika Savontaus
- Research Centre for Integrative Physiology & Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
- Turku Center for Disease Modeling, University of Turku, Turku, Finland
- Unit of Clinical Pharmacology, Turku University Hospital, Turku, Finland
| | - Risto Kerkelä
- Research Unit of Biomedicine and Internal Medicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Petteri Rinne
- Research Centre for Integrative Physiology & Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland.
- Turku Center for Disease Modeling, University of Turku, Turku, Finland.
| |
Collapse
|
6
|
Pohjolainen L, Kinnunen SM, Auno S, Kiriazis A, Pohjavaara S, Kari-Koskinen J, Zore M, Jumppanen M, Yli-Kauhaluoma J, Talman V, Ruskoaho H, Välimäki MJ. Switching of hypertrophic signalling towards enhanced cardiomyocyte identity and maturity by a GATA4-targeted compound. Stem Cell Res Ther 2024; 15:5. [PMID: 38167208 PMCID: PMC10763434 DOI: 10.1186/s13287-023-03623-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 12/20/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND The prevalence of heart failure is constantly increasing, and the prognosis of patients remains poor. New treatment strategies to preserve cardiac function and limit cardiac hypertrophy are therefore urgently needed. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are increasingly used as an experimental platform for cardiac in vitro studies. However, in contrast to adult cardiomyocytes, hiPSC-CMs display immature morphology, contractility, gene expression and metabolism and hence express a naive phenotype that resembles more of a foetal cardiomyocyte. METHODS A library of 14 novel compounds was synthesized in-house and screened for GATA4-NKX2-5 reporter activity and cellular toxicity. The most potent compound, 3i-1262, along with previously reported GATA4-acting compounds, were selected to investigate their effects on hypertrophy induced by endothelin-1 or mechanical stretch. Morphological changes and protein expression were characterized using immunofluorescence staining and high-content analysis. Changes in gene expression were studied using qPCR and RNA sequencing. RESULTS The prototype compound 3i-1262 inhibited GATA4-NKX2-5 synergy in a luciferase reporter assay. Additionally, the isoxazole compound 3i-1262 inhibited the hypertrophy biomarker B-type natriuretic peptide (BNP) by reducing BNP promoter activity and proBNP expression in neonatal rat ventricular myocytes and hiPSC-CMs, respectively. Treatment with 3i-1262 increased metabolic activity and cardiac troponin T expression in hiPSC-CMs without affecting GATA4 protein levels. RNA sequencing analysis revealed that 3i-1262 induces gene expression related to metabolic activity and cell cycle exit, indicating a change in the identity and maturity status of hiPSC-CMs. The biological processes that were enriched in upregulated genes in response to 3i-1262 were downregulated in response to mechanical stretch, and conversely, the downregulated processes in response to 3i-1262 were upregulated in response to mechanical stretch. CONCLUSIONS There is currently a lack of systematic understanding of the molecular modulation and control of hiPSC-CM maturation. In this study, we demonstrated that the GATA4-interfering compound 3i-1262 reorganizes the cardiac transcription factor network and converts hypertrophic signalling towards enhanced cardiomyocyte identity and maturity. This conceptually unique approach provides a novel structural scaffold for further development as a modality to promote cardiomyocyte specification and maturity.
Collapse
Affiliation(s)
- Lotta Pohjolainen
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland
| | - Sini M Kinnunen
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland
| | - Samuli Auno
- Drug Research Program and Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Alexandros Kiriazis
- Drug Research Program and Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Saana Pohjavaara
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland
| | - Julia Kari-Koskinen
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland
| | - Matej Zore
- Drug Research Program and Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Mikael Jumppanen
- Drug Research Program and Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Jari Yli-Kauhaluoma
- Drug Research Program and Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Virpi Talman
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland
| | - Heikki Ruskoaho
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland
| | - Mika J Välimäki
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland.
| |
Collapse
|
7
|
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
|
8
|
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: 13] [Impact Index Per Article: 6.5] [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
|
9
|
Pohjolainen L, Ruskoaho H, Talman V. Transcriptomics reveal stretched human pluripotent stem cell-derived cardiomyocytes as an advantageous hypertrophy model. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2022; 2:100020. [PMID: 39802492 PMCID: PMC11708431 DOI: 10.1016/j.jmccpl.2022.100020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 11/10/2022] [Accepted: 11/10/2022] [Indexed: 01/16/2025]
Abstract
Left ventricular hypertrophy, characterized by hypertrophy of individual cardiomyocytes, is an adaptive response to an increased cardiac workload that eventually leads to heart failure. Previous studies using neonatal rat ventricular myocytes (NRVMs) and animal models have revealed several genes and signaling pathways associated with hypertrophy and mechanical load. However, these models are not directly applicable to humans. Here, we studied the effect of cyclic mechanical stretch on gene expression of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) using RNA sequencing. hiPSC-CMs showed distinct hypertrophic changes in gene expression at the level of individual genes and in biological processes. We also identified several differentially expressed genes that have not been previously associated with cardiomyocyte hypertrophy and thus serve as attractive targets for future studies. When compared to previously published data attained from stretched NRVMs and human embryonic stem cell-derived cardiomyocytes, hiPSC-CMs displayed a smaller number of changes in gene expression, but the differentially expressed genes revealed more pronounced enrichment of hypertrophy-related biological processes and pathways. Overall, these results establish hiPSC-CMs as a valuable in vitro model for studying human cardiomyocyte hypertrophy.
Collapse
Affiliation(s)
- Lotta Pohjolainen
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Heikki Ruskoaho
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Virpi Talman
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| |
Collapse
|
10
|
Li Y, Wei L, Lan L, Gao Y, Zhang Q, Dawit H, Mao J, Guo L, Shen L, Wang L. Conductive biomaterials for cardiac repair: A review. Acta Biomater 2022; 139:157-178. [PMID: 33887448 DOI: 10.1016/j.actbio.2021.04.018] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/26/2021] [Accepted: 04/10/2021] [Indexed: 12/18/2022]
Abstract
Myocardial infarction (MI) is one of the fatal diseases in humans. Its incidence is constantly increasing annually all over the world. The problem is accompanied by the limited regenerative capacity of cardiomyocytes, yielding fibrous scar tissue formation. The propagation of electrical impulses in such tissue is severely hampered, negatively influencing the normal heart pumping function. Thus, reconstruction of the internal cardiac electrical connection is currently a major concern of myocardial repair. Conductive biomaterials with or without cell loading were extensively investigated to address this problem. This article introduces a detailed overview of the recent progress in conductive biomaterials and fabrication methods of conductive scaffolds for cardiac repair. After that, the advances in myocardial tissue construction in vitro by the restoration of intercellular communication and simulation of the dynamic electrophysiological environment are systematically reviewed. Furthermore, the latest trend in the study of cardiac repair in vivo using various conductive patches is summarized. Finally, we discuss the achievements and shortcomings of the existing conductive biomaterials and the properties of an ideal conductive patch for myocardial repair. We hope this review will help readers understand the importance and usefulness of conductive biomaterials in cardiac repair and inspire researchers to design and develop new conductive patches to meet the clinical requirements. STATEMENT OF SIGNIFICANCE: After myocardial infarction, the infarcted myocardial area is gradually replaced by heterogeneous fibrous tissue with inferior conduction properties, resulting in arrhythmia and heart remodeling. Conductive biomaterials have been extensively adopted to solve the problem. Summarizing the relevant literature, this review presents an overview of the types and fabrication methods of conductive biomaterials, and focally discusses the recent advances in myocardial tissue construction in vitro and myocardial repair in vivo, which is rarely covered in previous reviews. As well, the deficiencies of the existing conductive patches and their construction strategies for myocardial repair are discussed as well as the improving directions. Confidently, the readers of this review would appreciate advantages and current limitations of conductive biomaterials/patches in cardiac repair.
Collapse
Affiliation(s)
- Yimeng Li
- Key Laboratory of Textile Science & Technology of Ministry of Education and College of Textiles, Donghua University, Shanghai, 201620, China; Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology, Donghua University, Shanghai, 201620, China
| | - Leqian Wei
- Key Laboratory of Textile Science & Technology of Ministry of Education and College of Textiles, Donghua University, Shanghai, 201620, China; Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology, Donghua University, Shanghai, 201620, China
| | - Lizhen Lan
- Key Laboratory of Textile Science & Technology of Ministry of Education and College of Textiles, Donghua University, Shanghai, 201620, China; Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology, Donghua University, Shanghai, 201620, China
| | - Yaya Gao
- Key Laboratory of Textile Science & Technology of Ministry of Education and College of Textiles, Donghua University, Shanghai, 201620, China; Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology, Donghua University, Shanghai, 201620, China
| | - Qian Zhang
- Key Laboratory of Textile Science & Technology of Ministry of Education and College of Textiles, Donghua University, Shanghai, 201620, China; Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology, Donghua University, Shanghai, 201620, China
| | - Hewan Dawit
- Key Laboratory of Textile Science & Technology of Ministry of Education and College of Textiles, Donghua University, Shanghai, 201620, China; Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology, Donghua University, Shanghai, 201620, China
| | - Jifu Mao
- Key Laboratory of Textile Science & Technology of Ministry of Education and College of Textiles, Donghua University, Shanghai, 201620, China; Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology, Donghua University, Shanghai, 201620, China.
| | - Lamei Guo
- Key Laboratory of Textile Science & Technology of Ministry of Education and College of Textiles, Donghua University, Shanghai, 201620, China
| | - Li Shen
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
| | - Lu Wang
- Key Laboratory of Textile Science & Technology of Ministry of Education and College of Textiles, Donghua University, Shanghai, 201620, China; Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology, Donghua University, Shanghai, 201620, China
| |
Collapse
|
11
|
Majid QA, Orsolits B, Pohjolainen L, Kovács Z, Földes G, Talman V. Application of Human Induced Pluripotent Stem Cell Technology for Cardiovascular Regenerative Pharmacology. Methods Mol Biol 2022; 2454:163-196. [PMID: 33755910 DOI: 10.1007/7651_2021_369] [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] [Indexed: 06/12/2023]
Abstract
Cardiovascular diseases are one of the leading causes of mortality in the western world. Myocardial infarction is among the most prevalent and results in significant cell loss within the myocardium. Similarly, numerous drugs have been identified as having cardiotoxic side effects. The adult human heart is however unable to instigate an effective repair mechanism and regenerate the myocardium in response to such damage. This is in large part due to the withdrawal of cardiomyocytes (CMs) from the cell cycle. Thus, identifying, screening, and developing agents that could enhance the proliferative capacity of CMs holds great potential in cardiac regeneration. Human induced pluripotent stem cells (hiPSCs) and their cardiovascular derivatives are excellent tools in the search for such agents. This chapter outlines state-of-the art techniques for the two-dimensional differentiation and attainment of hiPSC-derived CMs and endothelial cells (ECs). Bioreactor systems and three-dimensional spheroids derived from hiPSC-cardiovascular derivatives are explored as platforms for drug discovery before focusing on relevant assays that can be employed to assess cell proliferation and viability.
Collapse
Affiliation(s)
- Qasim A Majid
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Barbara Orsolits
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Lotta Pohjolainen
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Zsófia Kovács
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Gábor Földes
- National Heart and Lung Institute, Imperial College London, London, UK.
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary.
| | - Virpi Talman
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
12
|
Tarvainen I, Nunn RC, Tuominen RK, Jäntti MH, Talman V. Protein Kinase A-Mediated Effects of Protein Kinase C Partial Agonist 5-(Hydroxymethyl)Isophthalate 1a3 in Colorectal Cancer Cells. J Pharmacol Exp Ther 2022; 380:54-62. [PMID: 34697230 DOI: 10.1124/jpet.121.000848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 10/14/2021] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer is the third most commonly occurring cancer in men and the second in women. The global burden of colorectal cancer is projected to increase to over 2 million new cases with over 1 million deaths within the next 10 years, and there is a great need for new compounds with novel mechanisms of action. Our group has developed protein kinase C (PKC)-modulating isophthalic acid derivatives that induce cytotoxicity toward human cervical and prostate cancer cell lines. In this study, we investigated the effects of 5-(hydroxymethyl)isophthalate 1a3 (HMI-1a3) on colorectal cancer cell lines (Caco-2, Colo205, and HT29). HMI-1a3 inhibited cell proliferation, decreased cell viability, and induced an apoptotic response in all studied cell lines. These effects, however, were independent of PKC. Using serine/threonine kinome profiling and pharmacological kinase inhibitors, we identified activation of the cAMP/PKA pathway as a new mechanism of action for HMI-1a3-induced anticancer activity in colorectal cancer cell lines. Our current results strengthen the hypothesis for HMI-1a3 as a potential anticancer agent against various malignancies. SIGNIFICANCE STATEMENT: Colorectal cancer (CRC) is a common solid organ malignancy. This study demonstrates that the protein kinase C (PKC)-C1 domain-targeted isophthalatic acid derivative 5-(hydroxymethyl)isophthalate 1a3 (HMI-1a3) has anticancer activity on CRC cell lines independently of PKC. We identified PKA activation as a mechanism of HMI-1a3-induced anticancer effects. The results reveal a new anticancer mechanism of action for the partial PKC agonist HMI-1a3 and thus provide new insights for the development of PKC and PKA modulators for cancer therapy.
Collapse
Affiliation(s)
- Ilari Tarvainen
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Finland (I.T., R.C.N., R.K.T., M.H.J., V.T.)
| | - Rebecca C Nunn
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Finland (I.T., R.C.N., R.K.T., M.H.J., V.T.)
| | - Raimo K Tuominen
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Finland (I.T., R.C.N., R.K.T., M.H.J., V.T.)
| | - Maria H Jäntti
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Finland (I.T., R.C.N., R.K.T., M.H.J., V.T.)
| | - Virpi Talman
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Finland (I.T., R.C.N., R.K.T., M.H.J., V.T.)
| |
Collapse
|
13
|
Välimäki MJ, Leigh RS, Kinnunen SM, March AR, de Sande AH, Kinnunen M, Varjosalo M, Heinäniemi M, Kaynak BL, Ruskoaho H. GATA-targeted compounds modulate cardiac subtype cell differentiation in dual reporter stem cell line. Stem Cell Res Ther 2021; 12:190. [PMID: 33736688 PMCID: PMC7977156 DOI: 10.1186/s13287-021-02259-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 03/01/2021] [Indexed: 12/15/2022] Open
Abstract
Background Pharmacological modulation of cell fate decisions and developmental gene regulatory networks holds promise for the treatment of heart failure. Compounds that target tissue-specific transcription factors could overcome non-specific effects of small molecules and lead to the regeneration of heart muscle following myocardial infarction. Due to cellular heterogeneity in the heart, the activation of gene programs representing specific atrial and ventricular cardiomyocyte subtypes would be highly desirable. Chemical compounds that modulate atrial and ventricular cell fate could be used to improve subtype-specific differentiation of endogenous or exogenously delivered progenitor cells in order to promote cardiac regeneration. Methods Transcription factor GATA4-targeted compounds that have previously shown in vivo efficacy in cardiac injury models were tested for stage-specific activation of atrial and ventricular reporter genes in differentiating pluripotent stem cells using a dual reporter assay. Chemically induced gene expression changes were characterized by qRT-PCR, global run-on sequencing (GRO-seq) and immunoblotting, and the network of cooperative proteins of GATA4 and NKX2-5 were further explored by the examination of the GATA4 and NKX2-5 interactome by BioID. Reporter gene assays were conducted to examine combinatorial effects of GATA-targeted compounds and bromodomain and extraterminal domain (BET) inhibition on chamber-specific gene expression. Results GATA4-targeted compounds 3i-1000 and 3i-1103 were identified as differential modulators of atrial and ventricular gene expression. More detailed structure-function analysis revealed a distinct subclass of GATA4/NKX2-5 inhibitory compounds with an acetyl lysine-like domain that contributed to ventricular cells (%Myl2-eGFP+). Additionally, BioID analysis indicated broad interaction between GATA4 and BET family of proteins, such as BRD4. This indicated the involvement of epigenetic modulators in the regulation of GATA-dependent transcription. In this line, reporter gene assays with combinatorial treatment of 3i-1000 and the BET bromodomain inhibitor (+)-JQ1 demonstrated the cooperative role of GATA4 and BRD4 in the modulation of chamber-specific cardiac gene expression. Conclusions Collectively, these results indicate the potential for therapeutic alteration of cell fate decisions and pathological gene regulatory networks by GATA4-targeted compounds modulating chamber-specific transcriptional programs in multipotent cardiac progenitor cells and cardiomyocytes. The compound scaffolds described within this study could be used to develop regenerative strategies for myocardial regeneration. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02259-z.
Collapse
Affiliation(s)
- Mika J Välimäki
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014, Helsinki, Finland
| | - Robert S Leigh
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014, Helsinki, Finland
| | - Sini M Kinnunen
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014, Helsinki, Finland
| | - Alexander R March
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014, Helsinki, Finland
| | - Ana Hernández de Sande
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Matias Kinnunen
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland.,Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Markku Varjosalo
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland.,Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Merja Heinäniemi
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Bogac L Kaynak
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014, Helsinki, Finland.
| | - Heikki Ruskoaho
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014, Helsinki, Finland.
| |
Collapse
|
14
|
Mokosch AS, Gerbig S, Grevelding CG, Haeberlein S, Spengler B. High-resolution AP-SMALDI MSI as a tool for drug imaging in Schistosoma mansoni. Anal Bioanal Chem 2021; 413:2755-2766. [PMID: 33723627 PMCID: PMC8007518 DOI: 10.1007/s00216-021-03230-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/31/2020] [Accepted: 02/09/2021] [Indexed: 12/21/2022]
Abstract
Schistosoma mansoni is a parasitic flatworm causing schistosomiasis, an infectious disease affecting several hundred million people worldwide. Schistosomes live dioeciously, and upon pairing with the male, the female starts massive egg production, which causes pathology. Praziquantel (PZQ) is the only drug used, but it has an inherent risk of resistance development. Therefore, alternatives are needed. In the context of drug repurposing, the cancer drug imatinib was tested, showing high efficacy against S. mansoni in vitro. Besides the gonads, imatinib mainly affected the integrity of the intestine in males and females. In this study, we investigated the potential uptake and distribution of imatinib in adult schistosomes including its distribution kinetics. To this end, we applied for the first time atmospheric-pressure scanning microprobe matrix-assisted laser desorption/ionization mass spectrometry imaging (AP-SMALDI MSI) for drug imaging in paired S. mansoni. Our results indicate that imatinib was present in the esophagus and intestine of the male as early as 20 min after in vitro exposure, suggesting an oral uptake route. After one hour, the drug was also found inside the paired female. The detection of the main metabolite, N-desmethyl imatinib, indicated metabolization of the drug. Additionally, a marker signal for the female ovary was successfully applied to facilitate further conclusions regarding organ tropism of imatinib. Our results demonstrate that AP-SMALDI MSI is a useful method to study the uptake, tissue distribution, and metabolization of imatinib in S. mansoni. The results suggest using AP-SMALDI MSI also for investigating other antiparasitic compounds and their metabolites in schistosomes and other parasites. ![]()
Collapse
Affiliation(s)
- Annika S Mokosch
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University Giessen, 35392, Giessen, Germany
| | - Stefanie Gerbig
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University Giessen, 35392, Giessen, Germany
| | - Christoph G Grevelding
- Institute of Parasitology, BFS, Justus Liebig University Giessen, 35392, Giessen, Germany
| | - Simone Haeberlein
- Institute of Parasitology, BFS, Justus Liebig University Giessen, 35392, Giessen, Germany
| | - Bernhard Spengler
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University Giessen, 35392, Giessen, Germany.
| |
Collapse
|
15
|
Leigh RS, Ruskoaho HJ, Kaynak BL. Cholecystokinin peptide signaling is regulated by a TBX5-MEF2 axis in the heart. Peptides 2021; 136:170459. [PMID: 33249116 DOI: 10.1016/j.peptides.2020.170459] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 11/15/2022]
Abstract
The procholecystokinin (proCCK) gene encodes a secreted peptide known to regulate the digestive, endocrine, and nervous systems. Though recently proposed as a biomarker for heart dysfunction, its physiological role in both the embryonic and adult heart is poorly understood, and there are no reports of tissue-specific regulators of cholecystokinin signaling in the heart or other tissues. In the present study, mRNA of proCCK was observed in cardiac tissues during mouse embryonic development, establishing proCCK as an early marker of differentiated cardiomyocytes which is later restricted to anatomical subdomains of the neonatal heart. Three-dimensional analysis of the expression of proCCK and CCKAR/CCKBR receptors was performed using in situ hybridization and optical projection tomography, illustrating chamber-specific expression patterns in the postnatal heart. Transcription factor motif analyses indicated developmental cardiac transcription factors TBX5 and MEF2C as upstream regulators of proCCK, and this regulatory activity was confirmed in reporter gene assays. proCCK mRNA levels were also measured in the infarcted heart and in response to cyclic mechanical stretch and endothelin-1, indicating dynamic transcriptional regulation which might be leveraged for improved biomarker development. Functional analyses of exogenous cholecystokinin octapeptide (CCK-8) administration were performed in differentiating mouse embryonic stem cells (mESCs), and the results suggest that CCK-8 does not act as a differentiation modulator of cardiomyocyte subtypes. Collectively, these findings indicate that proCCK is regulated at the transcriptional level by TBX5-MEF2 and neurohormonal signaling, informing use of proCCK as a biomarker and future strategies for upstream manipulation of cholecystokinin signaling in the heart and other tissues.
Collapse
Affiliation(s)
- Robert S Leigh
- Drug Research Programme, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Heikki J Ruskoaho
- Drug Research Programme, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Bogac L Kaynak
- Drug Research Programme, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
16
|
Karhu ST, Ruskoaho H, Talman V. Distinct Regulation of Cardiac Fibroblast Proliferation and Transdifferentiation by Classical and Novel Protein Kinase C Isoforms: Possible Implications for New Antifibrotic Therapies. Mol Pharmacol 2021; 99:104-113. [PMID: 33239332 DOI: 10.1124/molpharm.120.000094] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 11/16/2020] [Indexed: 11/22/2022] Open
Abstract
Cardiac fibrosis is characterized by accumulation and activation of fibroblasts and excessive production of extracellular matrix, which results in myocardial stiffening and eventually leads to heart failure. Although previous work suggests that protein kinase C (PKC) isoforms play a role in cardiac fibrosis and remodeling, the results are conflicting. Moreover, the potential of targeting PKC with pharmacological tools to inhibit pathologic fibrosis has not been fully evaluated. Here we investigated the effects of selected PKC agonists and inhibitors on cardiac fibroblast (CF) phenotype, proliferation, and gene expression using primary adult mouse CFs, which spontaneously transdifferentiate into myofibroblasts in culture. A 48-hour exposure to the potent PKC activator phorbol 12-myristate 13-acetate (PMA) at 10 nM concentration reduced the intensity of α-smooth muscle actin staining by 56% and periostin mRNA levels by 60% compared with control. The decreases were inhibited with the pan-PKC inhibitor Gö6983 and the inhibitor of classical PKC isoforms Gö6976, suggesting that classical PKCs regulate CF transdifferentiation. PMA also induced a 33% decrease in 5-bromo-2'-deoxyuridine-positive CFs, which was inhibited with Gö6983 but not with Gö6976, indicating that novel PKC isoforms (nPKCs) regulate CF proliferation. Moreover, PMA downregulated the expression of collagen-encoding genes Col1a1 and Col3a1 nPKC-dependently, showing that PKC activation attenuates matrix synthesis in CFs. The partial PKC agonist isophthalate derivative bis(1-ethylpentyl) 5-(hydroxymethyl)isophthalate induced parallel changes in phenotype, cell cycle activity, and gene expression. In conclusion, our results reveal distinct PKC-dependent regulation of CF transdifferentiation and proliferation and suggest that PKC agonists exhibit potential as an antifibrotic treatment. SIGNIFICANCE STATEMENT: Cardiac fibrosis is a pathological process that contributes to the development of heart failure. The molecular mechanisms regulating fibrosis in the heart are, however, not fully understood, which hinders the development of new therapies. Here, we demonstrate that classical and novel protein kinase C (PKC) isoforms distinctly regulate cardiac fibroblast transdifferentiation and proliferation, the two central processes in fibrosis. Our results indicate that pharmacological PKC activation may be a promising strategy to inhibit myocardial fibrosis.
Collapse
Affiliation(s)
- S Tuuli Karhu
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Finland
| | - Heikki Ruskoaho
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Finland
| | - Virpi Talman
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Finland
| |
Collapse
|
17
|
Pohjolainen L, Easton J, Solanki R, Ruskoaho H, Talman V. Pharmacological Protein Kinase C Modulators Reveal a Pro-hypertrophic Role for Novel Protein Kinase C Isoforms in Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes. Front Pharmacol 2021; 11:553852. [PMID: 33584253 PMCID: PMC7874215 DOI: 10.3389/fphar.2020.553852] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 12/07/2020] [Indexed: 12/19/2022] Open
Abstract
Background: Hypertrophy of cardiomyocytes (CMs) is initially a compensatory mechanism to cardiac overload, but when prolonged, it leads to maladaptive myocardial remodeling, impairing cardiac function and causing heart failure. A key signaling molecule involved in cardiac hypertrophy is protein kinase C (PKC). However, the role of different PKC isoforms in mediating the hypertrophic response remains controversial. Both classical (cPKC) and novel (nPKC) isoforms have been suggested to play a critical role in rodents, whereas the role of PKC in hypertrophy of human CMs remains to be determined. Here, we aimed to investigate the effects of two different types of PKC activators, the isophthalate derivative HMI-1b11 and bryostatin-1, on CM hypertrophy and to elucidate the role of cPKCs and nPKCs in endothelin-1 (ET-1)-induced hypertrophy in vitro. Methods and Results: We used neonatal rat ventricular myocytes (NRVMs) and human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) to study the effects of pharmacological PKC modulators and ET-1. We used quantitative reverse transcription PCR to quantify hypertrophic gene expression and high-content analysis (HCA) to investigate CM morphology. In both cell types, ET-1, PKC activation (bryostatin-1 and HMI-1b11) and inhibition of cPKCs (Gö6976) increased hypertrophic gene expression. In NRVMs, these treatments also induced a hypertrophic phenotype as measured by increased recognition, intensity and area of α-actinin and F-actin fibers. Inhibition of all PKC isoforms with Gö6983 inhibited PKC agonist-induced hypertrophy, but could not fully block ET-1-induced hypertrophy. The mitogen-activated kinase kinase 1/2 inhibitor U0126 inhibited PKC agonist-induced hypertrophy fully and ET-1-induced hypertrophy partially. While ET-1 induced a clear increase in the percentage of pro-B-type natriuretic peptide-positive hiPSC-CMs, none of the phenotypic parameters used in HCA directly correlated with gene expression changes or with phenotypic changes observed in NRVMs. Conclusion: This work shows similar hypertrophic responses to PKC modulators in NRVMs and hiPSC-CMs. Pharmacological PKC activation induces CM hypertrophy via activation of novel PKC isoforms. This pro-hypertrophic effect of PKC activators should be considered when developing PKC-targeted compounds for e.g. cancer or Alzheimer’s disease. Furthermore, this study provides further evidence on distinct PKC-independent mechanisms of ET-1-induced hypertrophy both in NRVMs and hiPSC-CMs.
Collapse
Affiliation(s)
- Lotta Pohjolainen
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Julia Easton
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Reesha Solanki
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Heikki Ruskoaho
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Virpi Talman
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| |
Collapse
|
18
|
Piironen K, Haapala M, Talman V, Järvinen P, Sikanen T. Cell adhesion and proliferation on common 3D printing materials used in stereolithography of microfluidic devices. LAB ON A CHIP 2020; 20:2372-2382. [PMID: 32500123 DOI: 10.1039/d0lc00114g] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Three-dimensional (3D) printing has recently emerged as a cost-effective alternative for rapid prototyping of microfluidic devices. The feature resolution of stereolithography-based 3D printing is particularly well suited for manufacturing of continuous flow cell culture platforms. Poor cell adhesion or material-induced cell death may, however, limit the introduction of new materials to microfluidic cell culture. In this work, we characterized four commercially available materials commonly used in stereolithography-based 3D printing with respect to long-term (2 month) cell survival on native 3D printed surfaces. Cell proliferation rates, along with material-induced effects on apoptosis and cell survival, were examined in mouse embryonic fibroblasts. Additionally, the feasibility of Dental SG (material with the most favored properties) for culturing of human hepatocytes and human-induced pluripotent stem cells was evaluated. The strength of cell adhesion to Dental SG was further examined over a shear force gradient of 1-89 dyne per cm2 by using a custom-designed microfluidic shear force assay incorporating a 3D printed, tilted and tapered microchannel sealed with a polydimethylsiloxane lid. According to our results, autoclavation of the devices prior to cell seeding played the most important role in facilitating long-term cell survival on the native 3D printed surfaces with the shear force threshold in the range of 3-8 dyne per cm2.
Collapse
Affiliation(s)
- Kati Piironen
- Faculty of Pharmacy, Drug Research Program, Division of Pharmaceutical Chemistry and Technology, University of Helsinki, Viikinkaari 5E, FI-00014, Finland.
| | | | | | | | | |
Collapse
|
19
|
Sun G, Teng Y, Zhao Z, Cheow LF, Yu H, Chen CH. Functional Stem Cell Sorting via Integrative Droplet Synchronization. Anal Chem 2020; 92:7915-7923. [DOI: 10.1021/acs.analchem.0c01312] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Guoyun Sun
- Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, 04-08, Singapore
| | - Yao Teng
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 2 Medical Drive, MD9, Singapore
| | - Zixuan Zhao
- Mechanobiology Institute, National University of Singapore, 5A Engineering Drive 1, 04-08 Singapore
| | - Lih Feng Cheow
- Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, 04-08, Singapore
| | - Hanry Yu
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 2 Medical Drive, MD9, Singapore
- Mechanobiology Institute, National University of Singapore, 5A Engineering Drive 1, 04-08 Singapore
- Institute of Bioengineering and Nanotechnology, A*STAR, 31 Biopolis Way, The Nanos 07-01, Singapore
- CAMP, Singapore-MIT Alliance for Research and Technology, 1 CREATE Way, 04-01, Singapore
| | - Chia-Hung Chen
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong SAR China
| |
Collapse
|
20
|
GATA4-targeted compound exhibits cardioprotective actions against doxorubicin-induced toxicity in vitro and in vivo: establishment of a chronic cardiotoxicity model using human iPSC-derived cardiomyocytes. Arch Toxicol 2020; 94:2113-2130. [PMID: 32185414 PMCID: PMC7303099 DOI: 10.1007/s00204-020-02711-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 03/09/2020] [Indexed: 12/14/2022]
Abstract
Doxorubicin is a widely used anticancer drug that causes dose-related cardiotoxicity. The exact mechanisms of doxorubicin toxicity are still unclear, partly because most in vitro studies have evaluated the effects of short-term high-dose doxorubicin treatments. Here, we developed an in vitro model of long-term low-dose administration of doxorubicin utilizing human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Moreover, given that current strategies for prevention and management of doxorubicin-induced cardiotoxicity fail to prevent cancer patients developing heart failure, we also investigated whether the GATA4-targeted compound 3i-1000 has cardioprotective potential against doxorubicin toxicity both in vitro and in vivo. The final doxorubicin concentration used in the chronic toxicity model in vitro was chosen based on cell viability data evaluation. Exposure to doxorubicin at the concentrations of 1–3 µM markedly reduced (60%) hiPSC-CM viability already within 48 h, while a 14-day treatment with 100 nM doxorubicin concentration induced only a modest 26% reduction in hiPCS-CM viability. Doxorubicin treatment also decreased DNA content in hiPSC-CMs. Interestingly, the compound 3i-1000 attenuated doxorubicin-induced increase in pro-B-type natriuretic peptide (proBNP) expression and caspase-3/7 activation in hiPSC-CMs. Moreover, treatment with 3i-1000 for 2 weeks (30 mg/kg/day, i.p.) inhibited doxorubicin cardiotoxicity by restoring left ventricular ejection fraction and fractional shortening in chronic in vivo rat model. In conclusion, the results demonstrate that long-term exposure of hiPSC-CMs can be utilized as an in vitro model of delayed doxorubicin-induced toxicity and provide in vitro and in vivo evidence that targeting GATA4 may be an effective strategy to counteract doxorubicin-induced cardiotoxicity.
Collapse
|
21
|
Zanjanizadeh Ezazi N, Ajdary R, Correia A, Mäkilä E, Salonen J, Kemell M, Hirvonen J, Rojas OJ, Ruskoaho HJ, Santos HA. Fabrication and Characterization of Drug-Loaded Conductive Poly(glycerol sebacate)/Nanoparticle-Based Composite Patch for Myocardial Infarction Applications. ACS APPLIED MATERIALS & INTERFACES 2020; 12:6899-6909. [PMID: 31967771 PMCID: PMC7450488 DOI: 10.1021/acsami.9b21066] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Heart tissue engineering is critical in the treatment of myocardial infarction, which may benefit from drug-releasing smart materials. In this study, we load a small molecule (3i-1000) in new biodegradable and conductive patches for application in infarcted myocardium. The composite patches consist of a biocompatible elastomer, poly(glycerol sebacate) (PGS), coupled with collagen type I, used to promote cell attachment. In addition, polypyrrole is incorporated because of its electrical conductivity and to induce cell signaling. Results from the in vitro experiments indicate a high density of cardiac myoblast cells attached on the patches, which stay viable for at least 1 month. The degradation of the patches does not show any cytotoxic effect, while 3i-1000 delivery induces cell proliferation. Conductive patches show high blood wettability and drug release, correlating with the rate of degradation of the PGS matrix. Together with the electrical conductivity and elongation characteristics, the developed biomaterial fits the mechanical, conductive, and biological demands required for cardiac treatment.
Collapse
Affiliation(s)
- Nazanin Zanjanizadeh Ezazi
- Drug Research Program,
Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Rubina Ajdary
- Department of Bioproducts and Biosystems, School of Chemical
Engineering, Aalto University, P.O. Box 16300, FI-00076 Aalto, Espoo, Finland
| | - Alexandra Correia
- Drug Research Program,
Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Ermei Mäkilä
- Laboratory of Industrial Physics, Department of Physics and Astronomy, University of Turku, FI-20014 Turku, Finland
| | - Jarno Salonen
- Laboratory of Industrial Physics, Department of Physics and Astronomy, University of Turku, FI-20014 Turku, Finland
| | - Marianna Kemell
- Department of Chemistry, University of
Helsinki, FI-00014 Helsinki, Finland
| | - Jouni Hirvonen
- Drug Research Program,
Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Orlando J. Rojas
- Department of Bioproducts and Biosystems, School of Chemical
Engineering, Aalto University, P.O. Box 16300, FI-00076 Aalto, Espoo, Finland
- Departments of Chemical
& Biological Engineering, Chemistry, and Wood Science, The University of British Columbia, 2360 East Mall, Vancouver, British Columbia V6T 1Z3, Canada
| | - Heikki J. Ruskoaho
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Hélder A. Santos
- Drug Research Program,
Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
- Helsinki Institute of Life Science (HiLIFE), University of Helsinki, FI-00014 Helsinki, Finland
- E-mail: .
Tel: +358 2941 59661
| |
Collapse
|
22
|
Abstract
Various strategies have been applied to replace the loss of cardiomyocytes in order to restore reduced cardiac function and prevent the progression of heart disease. Intensive research efforts in the field of cellular reprogramming and cell transplantation may eventually lead to efficient in vivo applications for the treatment of cardiac injuries, representing a novel treatment strategy for regenerative medicine. Modulation of cardiac transcription factor (TF) networks by chemical entities represents another viable option for therapeutic interventions. Comprehensive screening projects have revealed a number of molecular entities acting on molecular pathways highly critical for cellular lineage commitment and differentiation, including compounds targeting Wnt- and transforming growth factor beta (TGFβ)-signaling. Furthermore, previous studies have demonstrated that GATA4 and NKX2-5 are essential TFs in gene regulation of cardiac development and hypertrophy. For example, both of these TFs are required to fully activate mechanical stretch-responsive genes such as atrial natriuretic peptide and brain natriuretic peptide (BNP). We have previously reported that the compound 3i-1000 efficiently inhibited the synergy of the GATA4-NKX2-5 interaction. Cellular effects of 3i-1000 have been further characterized in a number of confirmatory in vitro bioassays, including rat cardiac myocytes and animal models of ischemic injury and angiotensin II-induced pressure overload, suggesting the potential for small molecule-induced cardioprotection.
Collapse
Affiliation(s)
- Mika J. Välimäki
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of PharmacyUniversity of HelsinkiHelsinki, Finland
| | - Heikki J. Ruskoaho
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of PharmacyUniversity of HelsinkiHelsinki, Finland
| |
Collapse
|
23
|
Jumppanen M, Kinnunen SM, Välimäki MJ, Talman V, Auno S, Bruun T, Boije Af Gennäs G, Xhaard H, Aumüller IB, Ruskoaho H, Yli-Kauhaluoma J. Synthesis, Identification, and Structure-Activity Relationship Analysis of GATA4 and NKX2-5 Protein-Protein Interaction Modulators. J Med Chem 2019; 62:8284-8310. [PMID: 31431011 PMCID: PMC7076710 DOI: 10.1021/acs.jmedchem.9b01086] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
![]()
Transcription factors GATA4 and NKX2-5
directly interact and synergistically
activate several cardiac genes and stretch-induced cardiomyocyte hypertrophy.
Previously, we identified phenylisoxazole carboxamide 1 as a hit compound, which inhibited the GATA4–NKX2-5 transcriptional
synergy. Here, the chemical space around the molecular structure of 1 was explored by synthesizing and characterizing 220 derivatives
and structurally related compounds. In addition to the synergistic
transcriptional activation, selected compounds were evaluated for
their effects on transcriptional activities of GATA4 and NKX2-5 individually
as well as potential cytotoxicity. The structure–activity relationship
(SAR) analysis revealed that the aromatic isoxazole substituent in
the southern part regulates the inhibition of GATA4–NKX2-5
transcriptional synergy. Moreover, inhibition of GATA4 transcriptional
activity correlated with the reduced cell viability. In summary, comprehensive
SAR analysis accompanied by data analysis successfully identified
potent and selective inhibitors of GATA4–NKX2-5 transcriptional
synergy and revealed structural features important for it.
Collapse
Affiliation(s)
| | | | | | - Virpi Talman
- Imperial College London, Imperial Centre for Translational and Experimental Medicine , National Heart and Lung Institute , Du Cane Road , London W12 0NN , United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Bolt HM. Highlight report: Cell type selection for toxicity testing. EXCLI JOURNAL 2019; 17:1180-1181. [PMID: 30713478 PMCID: PMC6341425 DOI: 10.17179/excli2018-2017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 12/18/2018] [Indexed: 11/13/2022]
Affiliation(s)
- H M Bolt
- IfADo - Leibniz Research Centre for Working Environment and Human Factors, Dortmund, GERMANY
| |
Collapse
|