1
|
Tian R, Song H, Li J, Yuan T, Liu J, Wang Y, Li Y, Song X. PINCH-1 promotes tumor growth and metastasis by enhancing DRP1-mediated mitochondrial fission in head and neck squamous cell carcinoma. Cancer Biol Ther 2025; 26:2477365. [PMID: 40065703 PMCID: PMC11901378 DOI: 10.1080/15384047.2025.2477365] [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: 10/16/2024] [Revised: 02/25/2025] [Accepted: 03/05/2025] [Indexed: 03/14/2025] Open
Abstract
PURPOSE Abnormal expression of PINCH-1 has been observed in various types of human cancers. However, the clinical importance and mechanism underlying its role in head and neck squamous cell carcinoma (HNSCC) is yet to be fully elucidated. METHODS This study evaluated the expression of PINCH-1 in HNSCC samples through immunohistochemical staining and Western blotting. AMC-HN-8, Cal27, and SCC7 cell lines were utilized for cellular function experiments, both in vivo and in vitro. CCK8, colony-formation assay, flow cytometry, wound-healing assay, and transwell assay were employed to investigate the effects of alterations in target proteins on the growth and metastasis of cancer cells. Mito-Tracker Deep Red FM was used to track mitochondrial morphology. RESULTS PINCH-1 was found to be overexpressed in HNSCC and closely associated with lymph node metastasis and poor pathologic differentiation. Its upregulation promoted proliferation, inhibited apoptosis, and enhanced migration and invasion in HNSCC cells. It also promoted mitochondrial fission. We conducted a mechanism analysis, which showed that PINCH-1 knockdown inhibited mitochondrial fission by reducing the expression of DRP1. Furthermore, inhibition of mitochondrial fission could impede the proliferation and metastasis of HNSCC cells. Re-expression of DRP1 reversed the inhibitory effect of PINCH-1 knockdown on mitochondrial fission, cell proliferation, and metastasis in HNSCC cells. CONCLUSIONS PINCH-1 plays a critical oncogenic role in HNSCC by enhancing DRP1-mediated mitochondrial fission, which may serve as a novel therapeutic target for HNSCC.
Collapse
Affiliation(s)
- Ruxian Tian
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Hao Song
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Jiaxuan Li
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Ting Yuan
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Jiahui Liu
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Yaqi Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Yumei Li
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Xicheng Song
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong, China
| |
Collapse
|
2
|
Maxey AP, Travis JM, McCain ML. Regulation of oxytocin-induced calcium transients and gene expression in engineered myometrial tissues by tissue architecture and matrix rigidity. Curr Res Physiol 2023; 6:100108. [PMID: 38107790 PMCID: PMC10724203 DOI: 10.1016/j.crphys.2023.100108] [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: 05/30/2023] [Revised: 08/21/2023] [Accepted: 09/20/2023] [Indexed: 12/19/2023] Open
Abstract
The uterus is susceptible to benign tumors known as fibroids, which have been associated with many pregnancy complications, including preterm labor. However, the impact of fibrotic tissue remodeling on the physiology of the myometrium, the smooth muscle layer of the uterus, is poorly understood, in large part due to a lack of model systems. In this study, we engineered healthy-like and fibrotic-like myometrium by culturing human myometrial smooth muscle cells on polyacrylamide hydrogels micropatterned with fibronectin to independently tune matrix rigidity and tissue alignment, respectively. We then evaluated calcium transients in response to oxytocin stimulation. Isotropic myometrial tissues on stiff substrates (representing fibrotic myometrium) had shorter calcium transients due to shorter decay time compared to aligned myometrial tissues on soft substrates (representing healthy myometrium). Calcium transients in aligned tissues had longer response times and longer decay times than isotropic tissues, irrespective of substrate stiffness. The amplitude of calcium transients was also higher on soft substrates compared to stiff substrates, irrespective of tissue alignment. We also performed RNA sequencing to detect differentially expressed genes between healthy- and fibrotic-like tissues, which revealed that a bitter taste receptor shown to induce smooth muscle relaxation, TAS2R31, was down-regulated in fibrotic-like tissues. Finally, we measured oxytocin-induced calcium transients in response to pre-treatment with progesterone, caffeine, thrombin, and nifedipine to demonstrate applications for our model system in drug screening. Both progesterone and caffeine caused a decrease in calcium transient duration, as expected, while thrombin and nifedipine had less impact. Collectively, our engineered model of the myometrium enables new insights into myometrial mechanobiology and can be extended to identify or screen novel drug targets.
Collapse
Affiliation(s)
- Antonina P. Maxey
- Laboratory for Living Systems Engineering, Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | - Jaya M. Travis
- Laboratory for Living Systems Engineering, Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | - Megan L. McCain
- Laboratory for Living Systems Engineering, Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
3
|
Tani H, Kobayashi E, Yagi S, Tanaka K, Kameda-Haga K, Shibata S, Moritoki N, Takatsuna K, Moriwaki T, Sekine O, Umei TC, Morita Y, Soma Y, Kishino Y, Kanazawa H, Fujita J, Hattori S, Fukuda K, Tohyama S. Heart-derived collagen promotes maturation of engineered heart tissue. Biomaterials 2023; 299:122174. [PMID: 37285642 DOI: 10.1016/j.biomaterials.2023.122174] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 05/17/2023] [Accepted: 05/19/2023] [Indexed: 06/09/2023]
Abstract
Although the extracellular matrix (ECM) plays essential roles in heart tissue engineering, the optimal ECM components for heart tissue organization have not previously been elucidated. Here, we focused on the main ECM component, fibrillar collagen, and analyzed the effects of collagens on heart tissue engineering, by comparing the use of porcine heart-derived collagen and other organ-derived collagens in generating engineered heart tissue (EHT). We demonstrate that heart-derived collagen induces better contraction and relaxation of human induced pluripotent stem cell-derived EHT (hiPSC-EHT) and that hiPSC-EHT with heart-derived collagen exhibit more mature profiles than those with collagens from other organs. Further, we found that collagen fibril formation and gel stiffness influence the contraction, relaxation, and maturation of hiPSC-EHT, suggesting the importance of collagen types III and type V, which are relatively abundant in the heart. Thus, we demonstrate the effectiveness of organ-specific collagens in tissue engineering and drug discovery.
Collapse
Affiliation(s)
- Hidenori Tani
- Department of Cardiology, Japan; Joint Research Laboratory for Medical Innovation in Heart Disease, Japan
| | - Eiji Kobayashi
- Department of Organ Fabrication, Japan; Department of Kidney Regenerative Medicine, The Jikei University School of Medicine, Tokyo, Japan.
| | - Shinomi Yagi
- Nippi Research Institute of Biomatrix, Toride, Ibaraki, Japan
| | - Keisuke Tanaka
- Nippi Research Institute of Biomatrix, Toride, Ibaraki, Japan
| | | | - Shinsuke Shibata
- Electron Microscope Laboratory, Keio University School of Medicine, Shinjuku, Tokyo, Japan; Division of Microscopic Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Nobuko Moritoki
- Electron Microscope Laboratory, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | | | | | | | | | - Yuika Morita
- Department of Cardiology, Japan; Kanagawa Institute of Industrial Science and Technology (KISTEC), Kawasaki, Kanagawa, Japan
| | | | | | | | - Jun Fujita
- Department of Cardiology, Japan; Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Shunji Hattori
- Department of Kidney Regenerative Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | | | | |
Collapse
|
4
|
Middleton RC, Liao K, Liu W, de Couto G, Garcia N, Antes T, Wang Y, Wu D, Li X, Tourtellotte WG, Marbán E. Newt A1 cell-derived extracellular vesicles promote mammalian nerve growth. Sci Rep 2023; 13:11829. [PMID: 37481602 PMCID: PMC10363125 DOI: 10.1038/s41598-023-38671-z] [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: 04/11/2023] [Accepted: 07/12/2023] [Indexed: 07/24/2023] Open
Abstract
Newts have the extraordinary ability to fully regenerate lost or damaged cardiac, neural and retinal tissues, and even amputated limbs. In contrast, mammals lack these broad regenerative capabilities. While the molecular basis of newts' regenerative ability is the subject of active study, the underlying paracrine signaling factors involved remain largely uncharacterized. Extracellular vesicles (EVs) play an important role in cell-to-cell communication via EV cargo-mediated regulation of gene expression patterns within the recipient cells. Here, we report that newt myogenic precursor (A1) cells secrete EVs (A1EVs) that contain messenger RNAs associated with early embryonic development, neuronal differentiation, and cell survival. Exposure of rat primary superior cervical ganglion (SCG) neurons to A1EVs increased neurite outgrowth, facilitated by increases in mitochondrial respiration. Canonical pathway analysis pinpointed activation of NGF/ERK5 signaling in SCG neurons exposed to A1EV, which was validated experimentally. Thus, newt EVs drive neurite growth and complexity in mammalian primary neurons.
Collapse
Affiliation(s)
- Ryan C Middleton
- Smidt Heart Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd #2900A, Los Angeles, CA, 90048, USA
| | - Ke Liao
- Smidt Heart Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd #2900A, Los Angeles, CA, 90048, USA
| | - Weixin Liu
- Smidt Heart Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd #2900A, Los Angeles, CA, 90048, USA
| | - Geoff de Couto
- Smidt Heart Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd #2900A, Los Angeles, CA, 90048, USA
| | - Nahuel Garcia
- Gecorp, Av Juan Manuel de Rosas 899, San Miguel del Monte, Buenos Aires, Argentina
| | - Travis Antes
- Smidt Heart Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd #2900A, Los Angeles, CA, 90048, USA
| | - Yizhou Wang
- Applied Genomics, Computation and Translational Core, Cedars-Sinai Medical Center, 8700 Beverly Blvd #2900A, Los Angeles, CA, 90048, USA
| | - Di Wu
- Applied Genomics, Computation and Translational Core, Cedars-Sinai Medical Center, 8700 Beverly Blvd #2900A, Los Angeles, CA, 90048, USA
| | - Xinling Li
- Applied Genomics, Computation and Translational Core, Cedars-Sinai Medical Center, 8700 Beverly Blvd #2900A, Los Angeles, CA, 90048, USA
| | - Warren G Tourtellotte
- Department of Pathology, Cedars-Sinai Medical Center, 8700 Beverly Blvd #2900A, Los Angeles, CA, 90048, USA
| | - Eduardo Marbán
- Smidt Heart Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd #2900A, Los Angeles, CA, 90048, USA.
| |
Collapse
|
5
|
Guichard JL, Kane MS, Grenett M, Sandel M, Benavides GA, Bradley WE, Powell PC, Darley-Usmar V, Ballinger SW, Dell'Italia LJ. Mitochondrial haplotype modulates genome expression and mitochondrial structure/function in cardiomyocytes following volume overload. Am J Physiol Heart Circ Physiol 2023; 324:H484-H493. [PMID: 36800507 PMCID: PMC10010923 DOI: 10.1152/ajpheart.00371.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 02/07/2023] [Accepted: 02/07/2023] [Indexed: 02/19/2023]
Abstract
Mitochondrial DNA (mtDNA) haplotype regulates mitochondrial structure/function and reactive oxygen species in aortocaval fistula (ACF) in mice. Here, we unravel the mitochondrial haplotype effects on cardiomyocyte mitochondrial ultrastructure and transcriptome response to ACF in vivo. Phenotypic responses and quantitative transmission electron microscopy (TEM) and RNA sequence at 3 days were determined after sham surgery or ACF in vivo in cardiomyocytes from wild-type (WT) C57BL/6J (C57n:C57mt) and C3H/HeN (C3Hn:C3Hmt) and mitochondrial nuclear exchange mice (C57n:C3Hmt or C3Hn:C57mt). Quantitative TEM of cardiomyocyte mitochondria C3HWT hearts have more electron-dense compact mitochondrial cristae compared with C57WT. In response to ACF, mitochondrial area and cristae integrity are normal in C3HWT; however, there is mitochondrial swelling, cristae lysis, and disorganization in both C57WT and MNX hearts. Tissue analysis shows that C3HWT hearts have increased autophagy, antioxidant, and glucose fatty acid oxidation-related genes compared with C57WT. Comparative transcriptomic analysis of cardiomyocytes from ACF was dependent upon mtDNA haplotype. C57mtDNA haplotype was associated with increased inflammatory/protein synthesis pathways and downregulation of bioenergetic pathways, whereas C3HmtDNA showed upregulation of autophagy genes. In conclusion, ACF in vivo shows a protective response of C3Hmt haplotype that is in large part driven by mitochondrial nuclear genome interaction.NEW & NOTEWORTHY The results of this study support the effects of mtDNA haplotype on nuclear gene expression in cardiomyocytes. Currently, there is no acceptable therapy for volume overload due to mitral regurgitation. The findings of this study could suggest that mtDNA haplotype activates different pathways after ACF warrants further investigations on human population of heart disease from different ancestry backgrounds.
Collapse
Affiliation(s)
- Jason L Guichard
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Mariame Selma Kane
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
- Department of Veterans Affairs Medical Center, Birmingham, Alabama, United States
| | - Maximiliano Grenett
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Michael Sandel
- Wildlife, Fisheries, and Aquaculture, Mississippi State University, Starkville, Mississippi, United States
| | - Gloria A Benavides
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, United States
- UAB Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Wayne E Bradley
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Pamela Cox Powell
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Victor Darley-Usmar
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, United States
- UAB Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Scott W Ballinger
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, United States
- UAB Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Louis J Dell'Italia
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
- Department of Veterans Affairs Medical Center, Birmingham, Alabama, United States
| |
Collapse
|
6
|
Wu P, Sai X, Li Z, Ye X, Jin L, Liu G, Li G, Yang P, Zhao M, Zhu S, Liu N, Zhu P. Maturation of induced pluripotent stem cell-derived cardiomyocytes and its therapeutic effect on myocardial infarction in mouse. Bioact Mater 2023; 20:286-305. [PMID: 35702609 PMCID: PMC9167678 DOI: 10.1016/j.bioactmat.2022.05.024] [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: 03/19/2022] [Revised: 05/19/2022] [Accepted: 05/19/2022] [Indexed: 11/30/2022] Open
Abstract
Induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) have an irreplaceable role in the treatment of myocardial infarction (MI), which can be injected into the transplanted area with new cardiomyocytes (Cardiomyocytes, CMs), and improve myocardial function. However, the immaturity of the structure and function of iPSC-CMs is the main bottleneck at present. Since collagen participates in the formation of extracellular matrix (ECM), we synthesized nano colloidal gelatin (Gel) with collagen as the main component, and confirmed that the biomaterial has good biocompatibility and is suitable for cellular in vitro growth. Subsequently, we combined the PI3K/AKT/mTOR pathway inhibitor BEZ-235 with Gel and found that the two combined increased the sarcomere length and action potential amplitude (APA) of iPSC-CMs, and improved the Ca2+ processing ability, the maturation of mitochondrial morphological structure and metabolic function. Not only that, Gel can also prolong the retention rate of iPSC-CMs in the myocardium and increase the expression of Cx43 and angiogenesis in the transplanted area of mature iPSC-CMs, which also provides a reliable basis for the subsequent treatment of mature iPSC-CMs. BEZ-235 + Gel promotes the maturation of sarcomere structure in iPSC-CMs. BEZ-235 + Gel promotes electrophysiological maturation of iPSC-CMs. BEZ-235 + Gel increases mitochondrial respiration in iPSC-CMs. Gel loaded with mature iPSC-CMs enhanced angiogenesis and gap junction formation at the injection site.
Collapse
|
7
|
Guo T, Jiang CS, Yang SZ, Zhu Y, He C, Carter AB, Antony VB, Peng H, Zhou Y. Mitochondrial fission and bioenergetics mediate human lung fibroblast durotaxis. JCI Insight 2023; 8:e157348. [PMID: 36422990 PMCID: PMC9870082 DOI: 10.1172/jci.insight.157348] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 11/21/2022] [Indexed: 11/27/2022] Open
Abstract
Pulmonary fibrosis is characterized by stiffening of the extracellular matrix. Fibroblasts migrate in the direction of greater stiffness, a phenomenon termed durotaxis. The mechanically guided fibroblast migration could be a crucial step in the progression of lung fibrosis. In this study, we found primary human lung fibroblasts sense increasing matrix stiffness with a change of mitochondrial dynamics in favor of mitochondrial fission and increased production of ATP. Mitochondria polarize in the direction of a physiologically relevant stiffness gradient, with conspicuous localization to the leading edge, primarily lamellipodia and filopodia, of migrating lung fibroblasts. Matrix stiffness-regulated mitochondrial fission and durotactic lung fibroblast migration are mediated by a dynamin-related protein 1/mitochondrial fission factor-dependent (DRP1/MFF-dependent) pathway. Importantly, we found that the DRP1/MFF pathway is activated in fibrotic lung myofibroblasts in both human IPF and bleomycin-induced mouse lung fibrosis. These findings suggest that energy-producing mitochondria need to be sectioned via fission and repositioned in durotactic lung fibroblasts to meet the higher energy demand. This represents a potentially new mechanism through which mitochondria may contribute to the progression of fibrotic lung diseases. Inhibition of durotactic migration of lung fibroblasts may play an important role in preventing the progression of human idiopathic pulmonary fibrosis.
Collapse
Affiliation(s)
- Ting Guo
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Respiratory Medicine, the Second Xiangya Hospital, Central-South University, Changsha, China
| | - Chun-sun Jiang
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Shan-Zhong Yang
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Yi Zhu
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Chao He
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - A. Brent Carter
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Birmingham Veterans Administration Medical Center, Birmingham, Alabama, USA
| | - Veena B. Antony
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Hong Peng
- Department of Respiratory Medicine, the Second Xiangya Hospital, Central-South University, Changsha, China
| | - Yong Zhou
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
8
|
Cai L, Shi L, Peng Z, Sun Y, Chen J. Ageing of skeletal muscle extracellular matrix and mitochondria: finding a potential link. Ann Med 2023; 55:2240707. [PMID: 37643318 PMCID: PMC10732198 DOI: 10.1080/07853890.2023.2240707] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/13/2023] [Accepted: 07/21/2023] [Indexed: 08/31/2023] Open
Abstract
Aim: To discuss the progress of extracellular matrix (ECM) characteristics, mitochondrial homeostasis, and their potential crosstalk in the pathogenesis of sarcopenia, a geriatric syndrome characterized by a generalized and progressive reduction in muscle mass, strength, and physical performance.Methods: This review focuses on the anatomy and physiology of skeletal muscle, alterations of ECM and mitochondria during ageing, and the role of the interplay between ECM and mitochondria in the pathogenesis of sarcopenia.Results: Emerging evidence points to a clear interplay between mitochondria and ECM in various tissues and organs. Under the ageing process, the ECM undergoes changes in composition and physical properties that may mediate mitochondrial changes via the systematic metabolism, ROS, SPARC pathway, and AMPK/PGC-1α signalling, which in turn exacerbate muscle degeneration. However, the precise effects of such crosstalk on the pathobiology of ageing, particularly in skeletal muscle, have not yet been fully understood.Conclusion: The changes in skeletal muscle ECM and mitochondria are partially responsible for the worsened muscle function during the ageing process. A deeper understanding of their alterations and interactions in sarcopenic patients can help prevent sarcopenia and improve its prognoses.
Collapse
Affiliation(s)
- Lubing Cai
- Department of Sports Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Luze Shi
- Department of Sports Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhen Peng
- Department of Sports Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yaying Sun
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiwu Chen
- Department of Sports Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
9
|
Germain N, Dhayer M, Dekiouk S, Marchetti P. Current Advances in 3D Bioprinting for Cancer Modeling and Personalized Medicine. Int J Mol Sci 2022; 23:3432. [PMID: 35408789 PMCID: PMC8998835 DOI: 10.3390/ijms23073432] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/15/2022] [Accepted: 03/18/2022] [Indexed: 02/01/2023] Open
Abstract
Tumor cells evolve in a complex and heterogeneous environment composed of different cell types and an extracellular matrix. Current 2D culture methods are very limited in their ability to mimic the cancer cell environment. In recent years, various 3D models of cancer cells have been developed, notably in the form of spheroids/organoids, using scaffold or cancer-on-chip devices. However, these models have the disadvantage of not being able to precisely control the organization of multiple cell types in complex architecture and are sometimes not very reproducible in their production, and this is especially true for spheroids. Three-dimensional bioprinting can produce complex, multi-cellular, and reproducible constructs in which the matrix composition and rigidity can be adapted locally or globally to the tumor model studied. For these reasons, 3D bioprinting seems to be the technique of choice to mimic the tumor microenvironment in vivo as closely as possible. In this review, we discuss different 3D-bioprinting technologies, including bioinks and crosslinkers that can be used for in vitro cancer models and the techniques used to study cells grown in hydrogels; finally, we provide some applications of bioprinted cancer models.
Collapse
Affiliation(s)
- Nicolas Germain
- UMR 9020–UMR-S 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, Institut de Recherche Contre le Cancer de Lille, University Lille, CNRS, Inserm, CHU Lille, F-59000 Lille, France; (M.D.); (S.D.)
- Banque de Tissus, Centre de Biologie-Pathologie, CHU Lille, F-59000 Lille, France
| | - Melanie Dhayer
- UMR 9020–UMR-S 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, Institut de Recherche Contre le Cancer de Lille, University Lille, CNRS, Inserm, CHU Lille, F-59000 Lille, France; (M.D.); (S.D.)
| | - Salim Dekiouk
- UMR 9020–UMR-S 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, Institut de Recherche Contre le Cancer de Lille, University Lille, CNRS, Inserm, CHU Lille, F-59000 Lille, France; (M.D.); (S.D.)
| | - Philippe Marchetti
- UMR 9020–UMR-S 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, Institut de Recherche Contre le Cancer de Lille, University Lille, CNRS, Inserm, CHU Lille, F-59000 Lille, France; (M.D.); (S.D.)
- Banque de Tissus, Centre de Biologie-Pathologie, CHU Lille, F-59000 Lille, France
| |
Collapse
|
10
|
Yanes B, Rainero E. The Interplay between Cell-Extracellular Matrix Interaction and Mitochondria Dynamics in Cancer. Cancers (Basel) 2022; 14:1433. [PMID: 35326584 PMCID: PMC8946811 DOI: 10.3390/cancers14061433] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/01/2022] [Accepted: 03/08/2022] [Indexed: 01/27/2023] Open
Abstract
The tumor microenvironment, in particular the extracellular matrix (ECM), plays a pivotal role in controlling tumor initiation and progression. In particular, the interaction between cancer cells and the ECM promotes cancer cell growth and invasion, leading to the formation of distant metastasis. Alterations in cancer cell metabolism is a key hallmark of cancer, which is often associated with alterations in mitochondrial dynamics. Recent research highlighted that, changes in mitochondrial dynamics are associated with cancer migration and metastasis-these has been extensively reviewed elsewhere. However, less is known about the interplay between the extracellular matrix and mitochondria functions. In this review, we will highlight how ECM remodeling associated with tumorigenesis contribute to the regulation of mitochondrial function, ultimately promoting cancer cell metabolic plasticity, able to fuel cancer invasion and metastasis.
Collapse
Affiliation(s)
| | - Elena Rainero
- School of Biosciences, The University of Sheffield, Western Bank, Sheffield S10 2TN, UK;
| |
Collapse
|
11
|
Chang YW, Song ZH, Chen CC. FAK regulates cardiomyocyte mitochondrial fission and function through Drp1. FEBS J 2021; 289:1897-1910. [PMID: 34739186 DOI: 10.1111/febs.16263] [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: 03/04/2021] [Revised: 09/19/2021] [Accepted: 11/03/2021] [Indexed: 11/29/2022]
Abstract
Loss of the mitochondrial fission enzyme dynamin-related protein 1 (Drp1) in cardiomyocytes results in energy shortage and heart failure. We aim to understand the intracellular signal pathway and extracellular factors regulating Drp1 phosphorylation and mitochondrial morphology and function in cardiomyocytes. We found cyclic mechanical stretching induced mitochondrial fission through Drp1 and focal adhesion kinase (FAK) in neonatal rat ventricular myocytes (NRVMs). FAK regulated phosphorylation of Drp1 and mitochondrial Drp1 levels. Extracellular fibronectin activated Drp1 and caused mitochondrial fission through FAK and extracellular signal-regulated kinase 1/2 (ERK1/2). Fibronectin increased NRVMs oxygen consumption rate and ATP content via FAK-ERK1/2-Drp1. Inhibition of the FAK-ERK1/2-Drp1 pathway caused cellular energy shortage. In addition, the FAK-ERK1/2-Drp1 pathway was rapidly activated by adrenergic agonists and contributed to agonists-stimulated NRVMs respiration. Interestingly, fibronectin limited the adrenergic agonists-induced NRVMs respiration by restricting phosphorylation of Drp1. Our results suggest that extracellular fibronectin and adrenergic stimulations use the FAK-ERK1/2-Drp1 pathway to regulate mitochondrial morphology and function in cardiomyocytes.
Collapse
Affiliation(s)
- Yu-Wang Chang
- Institute of Biomedical Sciences, Academia Sinica, Taiwan
| | - Zong-Han Song
- Institute of Biomedical Sciences, Academia Sinica, Taiwan
| | | |
Collapse
|
12
|
Selman M, Pardo A. Fibroageing: An ageing pathological feature driven by dysregulated extracellular matrix-cell mechanobiology. Ageing Res Rev 2021; 70:101393. [PMID: 34139337 DOI: 10.1016/j.arr.2021.101393] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 06/04/2021] [Accepted: 06/11/2021] [Indexed: 02/06/2023]
Abstract
Ageing is a multifactorial biological process leading to a progressive decline of physiological functions. The process of ageing includes numerous changes in the cells and the interactions between cell-cell and cell-microenvironment remaining as a critical risk factor for the development of chronic degenerative diseases. Systemic inflammation, known as inflammageing, increases as a consequence of ageing contributing to age-related morbidities. But also, persistent and uncontrolled activation of fibrotic pathways, with excessive accumulation of extracellular matrix (ECM) and organ dysfunction is markedly more frequent in the elderly. In this context, we introduce here the concept of Fibroageing, that is, the propensity to develop tissue fibrosis associated with ageing, and propose that ECM is a key player underlying this process. During ageing, molecules of the ECM become damaged through many modifications including glycation, crosslinking, and accumulation, leading to matrix stiffness which intensifies ageing-associated alterations. We provide a framework with some mechanistic hypotheses proposing that stiff ECM, in addition to the well-known activation of fibrotic positive feedback loops, affect several of the hallmarks of ageing, such as cell senescence and mitochondrial dysfunction, and in this context, is a key mechanism and a driver thread of Fibroageing.
Collapse
|
13
|
Khalil NN, McCain ML. Engineering the Cellular Microenvironment of Post-infarct Myocardium on a Chip. Front Cardiovasc Med 2021; 8:709871. [PMID: 34336962 PMCID: PMC8316619 DOI: 10.3389/fcvm.2021.709871] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 06/14/2021] [Indexed: 01/02/2023] Open
Abstract
Myocardial infarctions are one of the most common forms of cardiac injury and death worldwide. Infarctions cause immediate necrosis in a localized region of the myocardium, which is followed by a repair process with inflammatory, proliferative, and maturation phases. This repair process culminates in the formation of scar tissue, which often leads to heart failure in the months or years after the initial injury. In each reparative phase, the infarct microenvironment is characterized by distinct biochemical, physical, and mechanical features, such as inflammatory cytokine production, localized hypoxia, and tissue stiffening, which likely each contribute to physiological and pathological tissue remodeling by mechanisms that are incompletely understood. Traditionally, simplified two-dimensional cell culture systems or animal models have been implemented to elucidate basic pathophysiological mechanisms or predict drug responses following myocardial infarction. However, these conventional approaches offer limited spatiotemporal control over relevant features of the post-infarct cellular microenvironment. To address these gaps, Organ on a Chip models of post-infarct myocardium have recently emerged as new paradigms for dissecting the highly complex, heterogeneous, and dynamic post-infarct microenvironment. In this review, we describe recent Organ on a Chip models of post-infarct myocardium, including their limitations and future opportunities in disease modeling and drug screening.
Collapse
Affiliation(s)
- Natalie N Khalil
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, United States
| | - Megan L McCain
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, United States.,Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
14
|
Maturation strategies and limitations of induced pluripotent stem cell-derived cardiomyocytes. Biosci Rep 2021; 41:226678. [PMID: 33057659 PMCID: PMC8209171 DOI: 10.1042/bsr20200833] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 10/06/2020] [Accepted: 10/12/2020] [Indexed: 12/14/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) have the ability to differentiate into cardiomyocytes (CMs). They are not only widely used in cardiac pharmacology screening, human heart disease modeling, and cell transplantation-based treatments, but also the most promising source of CMs for experimental and clinical applications. However, their use is largely restricted by the immature phenotype of structure and function, which is similar to embryonic or fetal CMs and has certain differences from adult CMs. In order to overcome this critical issue, many studies have explored and revealed new strategies to induce the maturity of iPSC-CMs. Therefore, this article aims to review recent induction methods of mature iPSC-CMs, related mechanisms, and limitations.
Collapse
|
15
|
Amit C, Sathe G, Shunmugam A, Athyala PK, Ghose V, Chitipothu S, Janakiraman N, Sundara R, Elchuri SV. Graphitic Carbon Nitride Causes Widespread Global Molecular Changes in Epithelial and Fibroblast Cells. ACS OMEGA 2021; 6:9368-9380. [PMID: 33869917 PMCID: PMC8047657 DOI: 10.1021/acsomega.0c05513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 03/23/2021] [Indexed: 06/12/2023]
Abstract
For scaffold and imaging applications, nanomaterials such as graphene and its derivatives have been widely used. Graphitic carbon nitride (g-C3N4) is among one such derivative of graphenes, which draws strong consideration due to its physicochemical properties and photocatalytic activity. To use g-C3N4 for biological applications, such as molecular imaging or drug delivery, it must interact with the epithelium, cross the epithelial barrier, and then come in contact with the extracellular matrix of the fibroblast cells. Thus, it becomes essential to understand its molecular mechanism of action. Hence, in this study, to understand the molecular reprogramming associated with g-C3N4, global gene expression using DNA microarrays and proteomics using tandem mass tag (TMT) labeling and mass spectrometry were performed in epithelial and fibroblast cells, respectively. Our results showed that g-C3N4 can cross the epithelial barrier by regulating the adherens junction proteins. Further, using g-C3N4-PDMS scaffolds as a mimic of the extracellular matrix for fibroblast cells, the common signaling pathways were identified between the epithelium and fibroblast cells. These pathways include Wnt signaling, integrin signaling, TGF-β signaling, cadherin signaling, oxidative stress response, ubiquitin proteasome pathway, and EGF receptor signaling pathways. These altered signature pathways identified could play a prominent role in g-C3N4-mediated cellular interactions in both epithelial and fibroblast cells. Additionally, β catenin, EGFR, and MAP2K2 protein-protein interaction networks could play a prominent role in fibroblast cell proliferation. The findings could further our knowledge on g-C3N4-mediated alterations in cellular molecular signatures, enabling the potential use of these materials for biological applications such as molecular imaging and drug delivery.
Collapse
Affiliation(s)
- Chatterjee Amit
- Department
of Nanobiotechnology, Vision Research Foundation, Chennai 600006, India
| | - Gajanan Sathe
- Institute
of Bioinformatics, Bangalore 560066, Karnataka, India
- Manipal
Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India
| | - Abinaya Shunmugam
- Department
of Physics, Indian Institute of Technology,
Madras, Chennai 600036, India
| | | | - Vivek Ghose
- Institute
of Bioinformatics, Bangalore 560066, Karnataka, India
| | - Srujana Chitipothu
- Central
Research Instrumentation Facility, Core Lab, Vision Research Foundation, Chennai 600006, India
| | | | - Ramaprabhu Sundara
- Department
of Physics, Indian Institute of Technology,
Madras, Chennai 600036, India
| | - Sailaja V. Elchuri
- Department
of Nanobiotechnology, Vision Research Foundation, Chennai 600006, India
| |
Collapse
|
16
|
Inbody SC, Sinquefield BE, Lewis JP, Horton RE. Biomimetic microsystems for cardiovascular studies. Am J Physiol Cell Physiol 2021; 320:C850-C872. [PMID: 33760660 DOI: 10.1152/ajpcell.00026.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Traditional tissue culture platforms have been around for several decades and have enabled key findings in the cardiovascular field. However, these platforms failed to recreate the mechanical and dynamic features found within the body. Organs-on-chips (OOCs) are cellularized microfluidic-based devices that can mimic the basic structure, function, and responses of organs. These systems have been successfully utilized in disease, development, and drug studies. OOCs are designed to recapitulate the mechanical, electrical, chemical, and structural features of the in vivo microenvironment. Here, we review cardiovascular-themed OOC studies, design considerations, and techniques used to generate these cellularized devices. Furthermore, we will highlight the advantages of OOC models over traditional cell culture vessels, discuss implementation challenges, and provide perspectives on the state of the field.
Collapse
Affiliation(s)
- Shelby C Inbody
- Cardiovascular Tissue Engineering Laboratory, Biomedical Engineering Department, Cullen College of Engineering, University of Houston, Houston, Texas
| | - Bridgett E Sinquefield
- Cardiovascular Tissue Engineering Laboratory, Biomedical Engineering Department, Cullen College of Engineering, University of Houston, Houston, Texas
| | - Joshua P Lewis
- Cardiovascular Tissue Engineering Laboratory, Biomedical Engineering Department, Cullen College of Engineering, University of Houston, Houston, Texas
| | - Renita E Horton
- Cardiovascular Tissue Engineering Laboratory, Biomedical Engineering Department, Cullen College of Engineering, University of Houston, Houston, Texas
| |
Collapse
|
17
|
Yip JK, Sarkar D, Petersen AP, Gipson JN, Tao J, Kale S, Rexius-Hall ML, Cho N, Khalil NN, Kapadia R, McCain ML. Contact photolithography-free integration of patterned and semi-transparent indium tin oxide stimulation electrodes into polydimethylsiloxane-based heart-on-a-chip devices for streamlining physiological recordings. LAB ON A CHIP 2021; 21:674-687. [PMID: 33439202 PMCID: PMC7968549 DOI: 10.1039/d0lc00948b] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Controlled electrical stimulation is essential for evaluating the physiology of cardiac tissues engineered in heart-on-a-chip devices. However, existing stimulation techniques, such as external platinum electrodes or opaque microelectrode arrays patterned on glass substrates, have limited throughput, reproducibility, or compatibility with other desirable features of heart-on-a-chip systems, such as the use of tunable culture substrates, imaging accessibility, or enclosure in a microfluidic device. In this study, indium tin oxide (ITO), a conductive, semi-transparent, and biocompatible material, was deposited onto glass and polydimethylsiloxane (PDMS)-coated coverslips as parallel or point stimulation electrodes using laser-cut tape masks. ITO caused substrate discoloration but did not prevent brightfield imaging. ITO-patterned substrates were microcontact printed with arrayed lines of fibronectin and seeded with neonatal rat ventricular myocytes, which assembled into aligned cardiac tissues. ITO deposited as parallel or point electrodes was connected to an external stimulator and used to successfully stimulate micropatterned cardiac tissues to generate calcium transients or propagating calcium waves, respectively. ITO electrodes were also integrated into the cantilever-based muscular thin film (MTF) assay to stimulate and quantify the contraction of micropatterned cardiac tissues. To demonstrate the potential for multiple ITO electrodes to be integrated into larger, multiplexed systems, two sets of ITO electrodes were deposited onto a single substrate and used to stimulate the contraction of distinct micropatterned cardiac tissues independently. Collectively, these approaches for integrating ITO electrodes into heart-on-a-chip devices are relatively facile, modular, and scalable and could have diverse applications in microphysiological systems of excitable tissues.
Collapse
Affiliation(s)
- Joycelyn K Yip
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA.
| | - Debarghya Sarkar
- Ming Hsieh Department of Electrical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA.
| | - Andrew P Petersen
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA.
| | - Jennifer N Gipson
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA.
| | - Jun Tao
- Ming Hsieh Department of Electrical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA.
| | - Salil Kale
- Ming Hsieh Department of Electrical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA.
| | - Megan L Rexius-Hall
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA.
| | - Nathan Cho
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA.
| | - Natalie N Khalil
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA.
| | - Rehan Kapadia
- Ming Hsieh Department of Electrical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA.
| | - Megan L McCain
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA. and Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
18
|
Sport Performance and Manual Therapies: A Review on the Effects on Mitochondrial, Sarcoplasmatic and Ca 2+ Flux Response. Healthcare (Basel) 2021; 9:healthcare9020181. [PMID: 33572071 PMCID: PMC7915302 DOI: 10.3390/healthcare9020181] [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: 12/23/2020] [Revised: 01/26/2021] [Accepted: 02/04/2021] [Indexed: 11/17/2022] Open
Abstract
The present narrative review aims to highlight the possible effects manual therapies could have on cells and mitochondria, as these effects could improve athletic performance management. To this aim, this review summarizes the relationship between mechanical stimulation, with a special focus on physical activity, and cell response based on the most recent mechanobiology findings. Mechanobiology analyzes how cells respond to mechanical stressors coming from the environment. Indeed, endogenous (e.g., blood pressure, heartbeat and gastrointestinal motility) and exogenous (e.g., physical activity and manual therapies) stimuli can induce biochemical and epigenetic modifications that alter protein synthesis with heavy consequences on cell behavior. Mechanical stress can also influence mitochondrial behavior (i.e., biogenesis, autophagy, fusion, fission and energy production), sarcoplasmic response and calcium ion (Ca2+) flux. Since manual therapies have been shown to affect the extracellular matrix, which represents a primary source of mechanical stress that may alter both the cytoskeleton and mitochondrial metabolism, it is conceivable manual therapies could also affect cellular and mitochondrial behavior. Lastly, by suggesting possible directions for future laboratory and clinical studies, the authors expect this review to inspire further research on how manual therapies could affect bioenergetic metabolism and, thus, athletic performance.
Collapse
|
19
|
Lyra-Leite DM, Petersen AP, Ariyasinghe NR, Cho N, McCain ML. Mitochondrial architecture in cardiac myocytes depends on cell shape and matrix rigidity. J Mol Cell Cardiol 2021; 150:32-43. [PMID: 33038389 PMCID: PMC11956898 DOI: 10.1016/j.yjmcc.2020.10.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 09/30/2020] [Accepted: 10/04/2020] [Indexed: 12/12/2022]
Abstract
Contraction of cardiac myocytes depends on energy generated by the mitochondria. During cardiac development and disease, the structure and function of the mitochondrial network in cardiac myocytes is known to remodel in concert with many other factors, including changes in nutrient availability, hemodynamic load, extracellular matrix (ECM) rigidity, cell shape, and maturation of other intracellular structures. However, the independent role of each of these factors on mitochondrial network architecture is poorly understood. In this study, we tested the hypothesis that cell aspect ratio (AR) and ECM rigidity regulate the architecture of the mitochondrial network in cardiac myocytes. To do this, we spin-coated glass coverslips with a soft, moderate, or stiff polymer. Next, we microcontact printed cell-sized rectangles of fibronectin with AR matching cardiac myocytes at various developmental or disease states onto the polymer surface. We then cultured neonatal rat ventricular myocytes on the patterned surfaces and used confocal microscopy and image processing techniques to quantify sarcomeric α-actinin volume, nucleus volume, and mitochondrial volume, surface area, and size distribution. On some substrates, α-actinin volume increased with cell AR but was not affected by ECM rigidity. Nucleus volume was mostly uniform across all conditions. In contrast, mitochondrial volume increased with cell AR on all substrates. Furthermore, mitochondrial surface area to volume ratio decreased as AR increased on all substrates. Large mitochondria were also more prevalent in cardiac myocytes with higher AR. For select AR, mitochondria were also smaller as ECM rigidity increased. Collectively, these results suggest that mitochondrial architecture in cardiac myocytes is strongly influenced by cell shape and moderately influenced by ECM rigidity. These data have important implications for understanding the factors that impact metabolic performance during heart development and disease.
Collapse
Affiliation(s)
- Davi M Lyra-Leite
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, United States of America; Center for Pharmacogenomics, Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States of America
| | - Andrew P Petersen
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, United States of America
| | - Nethika R Ariyasinghe
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, United States of America; Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States of America
| | - Nathan Cho
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, United States of America
| | - Megan L McCain
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, United States of America; Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA 90033, United States of America.
| |
Collapse
|
20
|
Chen K, Wang Y, Deng X, Guo L, Wu C. Extracellular matrix stiffness regulates mitochondrial dynamics through PINCH-1- and kindlin-2-mediated signalling. ACTA ACUST UNITED AC 2021. [DOI: 10.1016/j.crcbio.2021.100008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
21
|
Engineering Shape-Controlled Microtissues on Compliant Hydrogels with Tunable Rigidity and Extracellular Matrix Ligands. Methods Mol Biol 2020. [PMID: 33340354 DOI: 10.1007/978-1-0716-1174-6_5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2023]
Abstract
In vitro models that recapitulate key aspects of native tissue architecture and the physical microenvironment are emerging systems for modeling development and disease. For example, the myocardium consists of layers of aligned and coupled cardiac myocytes that are interspersed with supporting cells and embedded in a compliant extracellular matrix (ECM). These cell-cell and cell-matrix interactions are known to be important regulators of tissue physiology and pathophysiology. In this protocol, we describe a method for mimicking the alignment, cell-cell interactions, and rigidity of the myocardium by engineering an array of square, aligned cardiac microtissues on polyacrylamide hydrogels. This entails three key methods: (1) fabricating elastomer stamps with a microtissue pattern; (2) preparing polyacrylamide hydrogel culture substrates with tunable elastic moduli; and (3) transferring ECM proteins onto the surface of the hydrogels using microcontact printing. These hydrogels can then be seeded with cardiac myocytes or mixtures of cardiac myocytes and fibroblasts to adjust cell-cell interactions. Overall, this approach is advantageous because shape-controlled microtissues encompass both cell-cell and cell-matrix adhesions in a form factor that is relatively reproducible and scalable. Furthermore, polyacrylamide hydrogels are compatible with the traction force microscopy assay for quantifying contractility, a critical function of the myocardium. Although cardiac microtissues are the example presented in this protocol, the techniques are relatively versatile and could have many applications in modeling other tissue systems.
Collapse
|
22
|
Li J, Hua Y, Miyagawa S, Zhang J, Li L, Liu L, Sawa Y. hiPSC-Derived Cardiac Tissue for Disease Modeling and Drug Discovery. Int J Mol Sci 2020; 21:E8893. [PMID: 33255277 PMCID: PMC7727666 DOI: 10.3390/ijms21238893] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 11/18/2020] [Accepted: 11/18/2020] [Indexed: 12/20/2022] Open
Abstract
Relevant, predictive normal, or disease model systems are of vital importance for drug development. The difference between nonhuman models and humans could contribute to clinical trial failures despite ideal nonhuman results. As a potential substitute for animal models, human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (CMs) provide a powerful tool for drug toxicity screening, modeling cardiovascular diseases, and drug discovery. Here, we review recent hiPSC-CM disease models and discuss the features of hiPSC-CMs, including subtype and maturation and the tissue engineering technologies for drug assessment. Updates from the international multisite collaborators/administrations for development of novel drug discovery paradigms are also summarized.
Collapse
Affiliation(s)
- Junjun Li
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
- Department of Cell Design for Tissue Construction, Faculty of Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Ying Hua
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
| | - Jingbo Zhang
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
| | - Lingjun Li
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
| | - Li Liu
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
- Department of Design for Tissue Regeneration, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yoshiki Sawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
| |
Collapse
|
23
|
Guo L, Cui C, Wang J, Yuan J, Yang Q, Zhang P, Su W, Bao R, Ran J, Wu C. PINCH-1 regulates mitochondrial dynamics to promote proline synthesis and tumor growth. Nat Commun 2020; 11:4913. [PMID: 33004813 PMCID: PMC7529891 DOI: 10.1038/s41467-020-18753-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 09/11/2020] [Indexed: 12/26/2022] Open
Abstract
Reprograming of proline metabolism is critical for tumor growth. Here we show that PINCH-1 is highly expressed in lung adenocarcinoma and promotes proline synthesis through regulation of mitochondrial dynamics. Knockout (KO) of PINCH-1 increases dynamin-related protein 1 (DRP1) expression and mitochondrial fragmentation, which suppresses kindlin-2 mitochondrial translocation and interaction with pyrroline-5-carboxylate reductase 1 (PYCR1), resulting in inhibition of proline synthesis and cell proliferation. Depletion of DRP1 reverses PINCH-1 deficiency-induced defects on mitochondrial dynamics, proline synthesis and cell proliferation. Furthermore, overexpression of PYCR1 in PINCH-1 KO cells restores proline synthesis and cell proliferation, and suppresses DRP1 expression and mitochondrial fragmentation. Finally, ablation of PINCH-1 from lung adenocarcinoma in mouse increases DRP1 expression and inhibits PYCR1 expression, proline synthesis, fibrosis and tumor growth. Our results identify a signaling axis consisting of PINCH-1, DRP1 and PYCR1 that regulates mitochondrial dynamics and proline synthesis, and suggest an attractive strategy for alleviation of tumor growth.
Collapse
Affiliation(s)
- Ling Guo
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Academy for Advanced Interdisciplinary Studies and Department of Biology, Southern University of Science and Technology, Shenzhen, China.
| | - Chunhong Cui
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Academy for Advanced Interdisciplinary Studies and Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Jiaxin Wang
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Academy for Advanced Interdisciplinary Studies and Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Jifan Yuan
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Academy for Advanced Interdisciplinary Studies and Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Qingyang Yang
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Academy for Advanced Interdisciplinary Studies and Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Ping Zhang
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Academy for Advanced Interdisciplinary Studies and Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Wen Su
- Department of Pathology, Shenzhen University Health Science Center, Shenzhen, China
| | - Ruolu Bao
- Department of Pathology, Shenzhen University Health Science Center, Shenzhen, China
| | - Jingchao Ran
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Academy for Advanced Interdisciplinary Studies and Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Chuanyue Wu
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
24
|
Abstract
PURPOSE OF REVIEW In this article, we review the different model systems based on human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) and how they have been applied to identify the cardiotoxic effects of anticancer therapies. RECENT FINDINGS Developments on 2D and 3D culture systems enabled the use of hiPSC-CMs as screening platforms for cardiotoxic effects of anticancer therapies such as anthracyclines, monoclonal antibodies, and tyrosine kinase inhibitors. Combined with computational approaches and higher throughput screening technologies, they have also enabled mechanistic studies and the search for cardioprotective strategies. As the population ages and cancer treatments become more effective, the cardiotoxic effects of anticancer drugs become a bigger problem leading to an increased role of cardio-oncology. In the past decade, human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have become an important platform for preclinical drug tests, elucidating mechanisms of action for drugs, and identifying cardioprotective pathways that could be further explored in the development of combined treatments. In this article, we highlight 2D and 3D model systems based on hiPSC-CMs that have been used to study the cardiotoxic effects of anticancer drugs, investigating their mechanisms of action and the potential for patient-specific prediction. We also present some of the important challenges and opportunities in the field, indicating possible future developments and how they could impact the landscape of cardio-oncology.
Collapse
Affiliation(s)
- Davi M. Lyra-Leite
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, 320 E Superior St, Searle 8-525, Chicago, IL 60611, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, 320 E Superior St, Searle 8-525, Chicago, IL 60611, USA
| | - Paul W. Burridge
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, 320 E Superior St, Searle 8-525, Chicago, IL 60611, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, 320 E Superior St, Searle 8-525, Chicago, IL 60611, USA
| |
Collapse
|
25
|
Petersen AP, Cho N, Lyra-Leite DM, Santoso JW, Gupta D, Ariyasinghe NR, McCain ML. Regulation of calcium dynamics and propagation velocity by tissue microstructure in engineered strands of cardiac tissue. Integr Biol (Camb) 2020; 12:34-46. [PMID: 32118279 PMCID: PMC11956900 DOI: 10.1093/intbio/zyaa003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 01/27/2020] [Accepted: 01/30/2020] [Indexed: 01/13/2023]
Abstract
Disruptions to cardiac tissue microstructure are common in diseased or injured myocardium and are known substrates for arrhythmias. However, we have a relatively coarse understanding of the relationships between myocardial tissue microstructure, propagation velocity and calcium cycling, due largely to the limitations of conventional experimental tools. To address this, we used microcontact printing to engineer strands of cardiac tissue with eight different widths, quantified several structural and functional parameters and established correlation coefficients. As strand width increased, actin alignment, nuclei density, sarcomere index and cell aspect ratio decreased with unique trends. The propagation velocity of calcium waves decreased and the rise time of calcium transients increased with increasing strand width. The decay time constant of calcium transients decreased and then slightly increased with increasing strand width. Based on correlation coefficients, actin alignment was the strongest predictor of propagation velocity and calcium transient rise time. Sarcomere index and cell aspect ratio were also strongly correlated with propagation velocity. Actin alignment, sarcomere index and cell aspect ratio were all weak predictors of the calcium transient decay time constant. We also measured the expression of several genes relevant to propagation and calcium cycling and found higher expression of the genes that encode for connexin 43 (Cx43) and a subunit of L-type calcium channels in thin strands compared to isotropic tissues. Together, these results suggest that thinner strands have higher values of propagation velocity and calcium transient rise time due to a combination of favorable tissue microstructure and enhanced expression of genes for Cx43 and L-type calcium channels. These data are important for defining how microstructural features regulate intercellular and intracellular calcium handling, which is needed to understand mechanisms of propagation in physiological situations and arrhythmogenesis in pathological situations.
Collapse
Affiliation(s)
- Andrew P. Petersen
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA
| | - Nathan Cho
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA
| | - Davi M. Lyra-Leite
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA
| | - Jeffrey W. Santoso
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA
| | - Divya Gupta
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA
| | - Nethika R. Ariyasinghe
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA
| | - Megan L. McCain
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA
| |
Collapse
|
26
|
Sevoflurane Pre-conditioning Ameliorates Diabetic Myocardial Ischemia/Reperfusion Injury Via Differential Regulation of p38 and ERK. Sci Rep 2020; 10:23. [PMID: 31913350 PMCID: PMC6949279 DOI: 10.1038/s41598-019-56897-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 12/17/2019] [Indexed: 01/01/2023] Open
Abstract
Diabetes mellitus (DM) significantly increases myocardial ischemia/reperfusion (MI/R) injury. During DM, cardioprotection induced by conventional pre-conditioning (PreCon) is decreased due to impaired AMP-activated protein kinase (AMPK) signaling. The current study investigated whether PreCon with inhaled anesthetic sevoflurane (SF-PreCon) remains cardioprotective during DM, and identified the involved mechanisms. Normal diet (ND) and high-fat diet (HFD)-induced DM mice were randomized into control and SF-PreCon (3 cycles of 15-minute period exposures to 2% sevoflurane) groups before MI/R. SF-PreCon markedly reduced MI/R injury in DM mice, as evidenced by improved cardiac function (increased LVEF and ±Dp/dt), decreased infarct size, and decreased apoptosis. To determine the relevant role of AMPK, the effect of SF-PreCon was determined in cardiac-specific AMPKα2 dominant negative expressing mice (AMPK-DN). SF-PreCon decreased MI/R injury in AMPK-DN mice. To explore the molecular mechanisms responsible for SF-PreCon mediated cardioprotection in DM mice, cell survival molecules were screened. Interestingly, in ND mice, SF-PreCon significantly reduced MI/R-induced activation of p38, a pro-death MAPK, without altering ERK and JNK. In DM and AMPK-DN mice, the inhibitory effect of SF-PreCon upon p38 activation was significantly blunted. However, SF-PreCon significantly increased phosphorylation of ERK1/2, a pro-survival MAPK in DM and AMPK-DN mice. We demonstrate that SF-PreCon protects the heart via AMPK-dependent inhibition of pro-death MAPK in ND mice. However, SF-PreCon exerts cardioprotective action via AMPK-independent activation of a pro-survival MAPK member in DM mice. SF-PreCon may be beneficial compared to conventional PreCon in diabetes or clinical scenarios in which AMPK signaling is impaired.
Collapse
|
27
|
Lyra-Leite DM, Andres AM, Cho N, Petersen AP, Ariyasinghe NR, Kim SS, Gottlieb RA, McCain ML. Matrix-guided control of mitochondrial function in cardiac myocytes. Acta Biomater 2019; 97:281-295. [PMID: 31401347 PMCID: PMC6801042 DOI: 10.1016/j.actbio.2019.08.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 07/31/2019] [Accepted: 08/02/2019] [Indexed: 02/08/2023]
Abstract
In ventricular myocardium, extracellular matrix (ECM) remodeling is a hallmark of physiological and pathological growth, coincident with metabolic rewiring of cardiac myocytes. However, the direct impact of the biochemical and mechanical properties of the ECM on the metabolic function of cardiac myocytes is mostly unknown. Furthermore, understanding the impact of distinct biomaterials on cardiac myocyte metabolism is critical for engineering physiologically-relevant models of healthy and diseased myocardium. For these reasons, we systematically measured morphological and metabolic responses of neonatal rat ventricular myocytes cultured on fibronectin- or gelatin-coated polydimethylsiloxane (PDMS) of three elastic moduli and gelatin hydrogels with four elastic moduli. On all substrates, total protein content, cell morphology, and the ratio of mitochondrial DNA to nuclear DNA were preserved. Cytotoxicity was low on all substrates, although slightly higher on PDMS compared to gelatin hydrogels. We also quantified oxygen consumption rates and extracellular acidification rates using a Seahorse extracellular flux analyzer. Our data indicate that several metrics associated with baseline glycolysis and baseline and maximum mitochondrial function are enhanced when cardiac myocytes are cultured on gelatin hydrogels compared to all PDMS substrates, irrespective of substrate rigidity. These results yield new insights into how mechanical and biochemical cues provided by the ECM impact mitochondrial function in cardiac myocytes. STATEMENT OF SIGNIFICANCE: Cardiac development and disease are associated with remodeling of the extracellular matrix coincident with metabolic rewiring of cardiac myocytes. However, little is known about the direct impact of the biochemical and mechanical properties of the extracellular matrix on the metabolic function of cardiac myocytes. In this study, oxygen consumption rates were measured in neonatal rat ventricular myocytes maintained on several commonly-used biomaterial substrates to reveal new relationships between the extracellular matrix and cardiac myocyte metabolism. Several mitochondrial parameters were enhanced on gelatin hydrogels compared to synthetic PDMS substrates. These data are important for comprehensively understanding matrix-regulation of cardiac myocyte physiology. Additionally, these data should be considered when selecting scaffolds for engineering in vitro cardiac tissue models.
Collapse
Affiliation(s)
- Davi M Lyra-Leite
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles CA, 90089, United States
| | - Allen M Andres
- Smidt Heart Institute and Barbra Streisand Women's Heart Center, Cedars-Sinai Medical Center, Los Angeles CA, 90048, United States
| | - Nathan Cho
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles CA, 90089, United States
| | - Andrew P Petersen
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles CA, 90089, United States
| | - Nethika R Ariyasinghe
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles CA, 90089, United States
| | - Suyon Sarah Kim
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles CA, 90089, United States
| | - Roberta A Gottlieb
- Smidt Heart Institute and Barbra Streisand Women's Heart Center, Cedars-Sinai Medical Center, Los Angeles CA, 90048, United States
| | - Megan L McCain
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles CA, 90089, United States; Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles CA, 90033, United States.
| |
Collapse
|
28
|
Sparc, an EPS-induced gene, modulates the extracellular matrix and mitochondrial function via ILK/AMPK pathways in C2C12 cells. Life Sci 2019; 229:277-287. [DOI: 10.1016/j.lfs.2019.05.070] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 05/24/2019] [Accepted: 05/27/2019] [Indexed: 01/06/2023]
|
29
|
Hughes JH, Ewy JM, Chen J, Wong SY, Tharp KM, Stahl A, Kumar S. Transcriptomic analysis reveals that BMP4 sensitizes glioblastoma tumor-initiating cells to mechanical cues. Matrix Biol 2019; 85-86:112-127. [PMID: 31189077 DOI: 10.1016/j.matbio.2019.06.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 06/03/2019] [Accepted: 06/07/2019] [Indexed: 12/15/2022]
Abstract
The poor prognosis of glioblastoma (GBM) is associated with a highly invasive stem-like subpopulation of tumor-initiating cells (TICs), which drive recurrence and contribute to intra-tumoral heterogeneity through differentiation. These TICs are better able to escape extracellular matrix-imposed mechanical restrictions on invasion than their more differentiated progeny, and sensitization of TICs to extracellular matrix mechanics extends survival in preclinical models of GBM. However, little is known about the molecular basis of the relationship between TIC differentiation and mechanotransduction. Here we explore this relationship through a combination of transcriptomic analysis and studies with defined-stiffness matrices. We show that TIC differentiation induced by bone morphogenetic protein 4 (BMP4) suppresses expression of proteins relevant to extracellular matrix signaling and sensitizes TIC spreading to matrix stiffness. Moreover, our findings point towards a previously unappreciated connection between BMP4-induced differentiation, mechanotransduction, and metabolism. Notably, stiffness and differentiation modulate oxygen consumption, and inhibition of oxidative phosphorylation influences cell spreading in a stiffness- and differentiation-dependent manner. Our work integrates bioinformatic analysis with targeted molecular measurements and perturbations to yield new insight into how morphogen-induced differentiation influences how GBM TICs process mechanical inputs.
Collapse
Affiliation(s)
- Jasmine H Hughes
- UC Berkeley - UCSF Graduate Program in Bioengineering; Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Jeanette M Ewy
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Joseph Chen
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Sophie Y Wong
- UC Berkeley - UCSF Graduate Program in Bioengineering; Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Kevin M Tharp
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California San Francisco, San Francisco, CA 94720, USA
| | - Andreas Stahl
- UC Berkeley - UCSF Graduate Program in Bioengineering; Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Sanjay Kumar
- UC Berkeley - UCSF Graduate Program in Bioengineering; Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
30
|
Antoniou CK, Manolakou P, Magkas N, Konstantinou K, Chrysohoou C, Dilaveris P, Gatzoulis KA, Tousoulis D. Cardiac Resynchronisation Therapy and Cellular Bioenergetics: Effects Beyond Chamber Mechanics. Eur Cardiol 2019; 14:33-44. [PMID: 31131035 PMCID: PMC6523053 DOI: 10.15420/ecr.2019.2.2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cardiac resynchronisation therapy is a cornerstone in the treatment of advanced dyssynchronous heart failure. However, despite its widespread clinical application, precise mechanisms through which it exerts its beneficial effects remain elusive. Several studies have pointed to a metabolic component suggesting that, both in concert with alterations in chamber mechanics and independently of them, resynchronisation reverses detrimental changes to cellular metabolism, increasing energy efficiency and metabolic reserve. These actions could partially account for the existence of responders that improve functionally but not echocardiographically. This article will attempt to summarise key components of cardiomyocyte metabolism in health and heart failure, with a focus on the dyssynchronous variant. Both chamber mechanics-related and -unrelated pathways of resynchronisation effects on bioenergetics – stemming from the ultramicroscopic level – and a possible common underlying mechanism relating mechanosensing to metabolism through the cytoskeleton will be presented. Improved insights regarding the cellular and molecular effects of resynchronisation on bioenergetics will promote our understanding of non-response, optimal device programming and lead to better patient care.
Collapse
Affiliation(s)
| | - Panagiota Manolakou
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens Athens, Greece
| | - Nikolaos Magkas
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens Athens, Greece
| | - Konstantinos Konstantinou
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens Athens, Greece
| | - Christina Chrysohoou
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens Athens, Greece
| | - Polychronis Dilaveris
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens Athens, Greece
| | - Konstantinos A Gatzoulis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens Athens, Greece
| | - Dimitrios Tousoulis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens Athens, Greece
| |
Collapse
|
31
|
Brown GE, Khetani SR. Microfabrication of liver and heart tissues for drug development. Philos Trans R Soc Lond B Biol Sci 2019; 373:rstb.2017.0225. [PMID: 29786560 DOI: 10.1098/rstb.2017.0225] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/24/2017] [Indexed: 12/12/2022] Open
Abstract
Drug-induced liver- and cardiotoxicity remain among the leading causes of preclinical and clinical drug attrition, marketplace drug withdrawals and black-box warnings on marketed drugs. Unfortunately, animal testing has proven to be insufficient for accurately predicting drug-induced liver- and cardiotoxicity across many drug classes, likely due to significant differences in tissue functions across species. Thus, the field of in vitro human tissue engineering has gained increasing importance over the last 10 years. Technologies such as protein micropatterning, microfluidics, three-dimensional scaffolds and bioprinting have revolutionized in vitro platforms as well as increased the long-term phenotypic stability of both primary cells and stem cell-derived differentiated cells. Here, we discuss advances in engineering approaches for constructing in vitro human liver and heart models with utility for drug development. Design features and validation data of representative models are presented to highlight major trends followed by the discussion of pending issues. Overall, bioengineered liver and heart models have significantly advanced our understanding of organ function and injury, which will prove useful for mitigating the risk of drug-induced organ toxicity to human patients, reducing animal usage for preclinical drug testing, aiding in the discovery of novel therapeutics against human diseases, and ultimately for applications in regenerative medicine.This article is part of the theme issue 'Designer human tissue: coming to a lab near you'.
Collapse
Affiliation(s)
- Grace E Brown
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Salman R Khetani
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| |
Collapse
|
32
|
Kannan S, Kwon C. Regulation of cardiomyocyte maturation during critical perinatal window. J Physiol 2019; 598:2941-2956. [PMID: 30571853 DOI: 10.1113/jp276754] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 11/23/2018] [Indexed: 12/13/2022] Open
Abstract
A primary limitation in the use of pluripotent stem cell-derived cardiomyocytes (PSC-CMs) for both patient health and scientific investigation is the failure of these cells to achieve full functional maturity. In vivo, cardiomyocytes undergo numerous adaptive structural, functional and metabolic changes during maturation. By contrast, PSC-CMs fail to fully undergo these developmental processes, instead remaining arrested at an embryonic stage of maturation. There is thus a significant need to understand the biological processes underlying proper CM maturation in vivo. Here, we discuss what is known regarding the initiation and coordination of CM maturation. We postulate that there is a critical perinatal window, ranging from embryonic day 18.5 to postnatal day 14 in mice, in which the maturation process is exquisitely sensitive to perturbation. While the initiation mechanisms of this process are unknown, it is increasingly clear that maturation proceeds through interconnected regulatory circuits that feed into one another to coordinate concomitant structural, functional and metabolic CM maturation. We highlight PGC1α, SRF and the MEF2 family as transcription factors that may potentially mediate this cross-talk. We lastly discuss several emerging technologies that will facilitate future studies into the mechanisms of CM maturation. Further study will not only produce a better understanding of its key processes, but provide practical insights into developing a robust strategy to produce mature PSC-CMs.
Collapse
Affiliation(s)
- Suraj Kannan
- Johns Hopkins University School of Medicine, 733 North Broadway, Baltimore, MD, 21205, USA
| | - Chulan Kwon
- Johns Hopkins University School of Medicine, 733 North Broadway, Baltimore, MD, 21205, USA
| |
Collapse
|
33
|
Perestrelo T, Correia M, Ramalho-Santos J, Wirtz D. Metabolic and Mechanical Cues Regulating Pluripotent Stem Cell Fate. Trends Cell Biol 2018; 28:1014-1029. [DOI: 10.1016/j.tcb.2018.09.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 08/30/2018] [Accepted: 09/25/2018] [Indexed: 02/07/2023]
|
34
|
Rog-Zielinska EA, O'Toole ET, Hoenger A, Kohl P. Mitochondrial Deformation During the Cardiac Mechanical Cycle. Anat Rec (Hoboken) 2018; 302:146-152. [PMID: 30302911 PMCID: PMC6312496 DOI: 10.1002/ar.23917] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 04/22/2018] [Accepted: 05/11/2018] [Indexed: 12/17/2022]
Abstract
Cardiomyocytes both cause and experience continual cyclic deformation. The exact effects of this deformation on the properties of intracellular organelles are not well characterized, although they are likely to be relevant for cardiomyocyte responses to active and passive changes in their mechanical environment. In the present study we provide three‐dimensional ultrastructural evidence for mechanically induced mitochondrial deformation in rabbit ventricular cardiomyocytes over a range of sarcomere lengths representing myocardial tissue stretch, an unloaded “slack” state, and contracture. We also show structural indications for interaction of mitochondria with one another, as well as with other intracellular elements such as microtubules, sarcoplasmic reticulum and T‐tubules. The data presented here help to contextualize recent reports on the mechanosensitivity and cell‐wide connectivity of the mitochondrial network and provide a structural framework that may aide interpretation of mechanically‐regulated molecular signaling in cardiac cells. Anat Rec, 302:146–152, 2019. © 2018 The Authors. The Anatomical Record published by Wiley Periodicals, Inc. on behalf of American Association of Anatomists.
Collapse
Affiliation(s)
- E A Rog-Zielinska
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg, Bad Krozingen, and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - E T O'Toole
- Department of Molecular, Cellular and Developmental Biology, University of Colorado at Boulder, Boulder, Colorado, USA
| | - A Hoenger
- Department of Molecular, Cellular and Developmental Biology, University of Colorado at Boulder, Boulder, Colorado, USA
| | - P Kohl
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg, Bad Krozingen, and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
35
|
Ariyasinghe NR, Lyra-Leite DM, McCain ML. Engineering cardiac microphysiological systems to model pathological extracellular matrix remodeling. Am J Physiol Heart Circ Physiol 2018; 315:H771-H789. [PMID: 29906229 PMCID: PMC6230901 DOI: 10.1152/ajpheart.00110.2018] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 05/27/2018] [Accepted: 06/08/2018] [Indexed: 12/11/2022]
Abstract
Many cardiovascular diseases are associated with pathological remodeling of the extracellular matrix (ECM) in the myocardium. ECM remodeling is a complex, multifactorial process that often contributes to declines in myocardial function and progression toward heart failure. However, the direct effects of the many forms of ECM remodeling on myocardial cell and tissue function remain elusive, in part because conventional model systems used to investigate these relationships lack robust experimental control over the ECM. To address these shortcomings, microphysiological systems are now being developed and implemented to establish direct relationships between distinct features in the ECM and myocardial function with unprecedented control and resolution in vitro. In this review, we will first highlight the most prominent characteristics of ECM remodeling in cardiovascular disease and describe how these features can be mimicked with synthetic and natural biomaterials that offer independent control over multiple ECM-related parameters, such as rigidity and composition. We will then detail innovative microfabrication techniques that enable precise regulation of cellular architecture in two and three dimensions. We will also describe new approaches for quantifying multiple aspects of myocardial function in vitro, such as contractility, action potential propagation, and metabolism. Together, these collective technologies implemented as cardiac microphysiological systems will continue to uncover important relationships between pathological ECM remodeling and myocardial cell and tissue function, leading to new fundamental insights into cardiovascular disease, improved human disease models, and novel therapeutic approaches.
Collapse
Affiliation(s)
- Nethika R Ariyasinghe
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California , Los Angeles, California
| | - Davi M Lyra-Leite
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California , Los Angeles, California
| | - Megan L McCain
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California , Los Angeles, California
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California , Los Angeles, California
| |
Collapse
|
36
|
Edwards N, Langford-Smith AWW, Wilkinson FL, Alexander MY. Endothelial Progenitor Cells: New Targets for Therapeutics for Inflammatory Conditions With High Cardiovascular Risk. Front Med (Lausanne) 2018; 5:200. [PMID: 30042945 PMCID: PMC6048266 DOI: 10.3389/fmed.2018.00200] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 06/21/2018] [Indexed: 12/12/2022] Open
Abstract
Over the past decade, we have witnessed an exponential growth of interest into the role of endothelial progenitor cells (EPCs) in cardiovascular disease. While the major thinking revolves around EPC angiogenic repair properties, we have used a hypothesis-driven approach to discover disease-related defects in their characteristics and based on these findings, have identified opportunities for functional enhancement, which offer an exciting avenue for translation into clinical intervention. In this review, we focus on two groups; circulating myeloid angiogenic cells (MACs) and late outgrowth endothelial colony forming cells (ECFCs), and will discuss the unique properties and defects of each population, as new insights have been gained into the potential function of each sub-type using current techniques and multiomic technology. We will discuss their role in inflammatory disorders and alterations in mitochondrial function. In addition, we share key insights into the glycocalyx, and propose this network of membrane-bound proteoglycans and glycoproteins, covering the endothelium warrants further investigation in order to clarify its significance in ECFC regulation of vascularization and angiogenesis and ultimately for potential translational therapeutic aspects.
Collapse
Affiliation(s)
- Nicola Edwards
- Cardiovascular Science, Centre for Bioscience, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom
| | - Alexander W W Langford-Smith
- Cardiovascular Science, Centre for Bioscience, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom
| | - Fiona L Wilkinson
- Cardiovascular Science, Centre for Bioscience, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom
| | - M Yvonne Alexander
- Cardiovascular Science, Centre for Bioscience, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom.,Manchester Academic Health Science Centre, Manchester, United Kingdom
| |
Collapse
|
37
|
Microenvironmental Modulation of Calcium Wave Propagation Velocity in Engineered Cardiac Tissues. Cell Mol Bioeng 2018; 11:337-352. [PMID: 31719889 DOI: 10.1007/s12195-018-0522-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 04/09/2018] [Indexed: 10/17/2022] Open
Abstract
Introduction In the myocardium, rapid propagation of action potentials and subsequent calcium waves is critical for synchronizing the contraction of cardiac myocytes and maximizing cardiac output. In many pathological settings, diverse remodeling of the tissue microenvironment is correlated with arrhythmias and decreased cardiac output, but the precise impact of tissue remodeling on propagation is not completely understood. Our objective was to delineate how multiple features within the cardiac tissue microenvironment modulate propagation velocity. Methods To recapitulate diverse myocardial tissue microenvironments, we engineered substrates with tunable elasticity, patterning, composition, and topography using two formulations of polydimethylsiloxane (PDMS) micropatterned with fibronectin and gelatin hydrogels with flat or micromolded features. We cultured neonatal rat ventricular myocytes on these substrates and quantified cell density, tissue alignment, and cell shape. We used a fluorescent calcium indicator, high-speed microscopy, and newly-developed analysis software to record and quantify calcium wave propagation velocity (CPV). Results For all substrates, tissue alignment and cell aspect ratio were higher in aligned compared to isotropic tissues. Isotropic CPV and longitudinal CPV were similar across conditions, but transverse CPV was lower on micromolded gelatin hydrogels compared to micropatterned soft and stiff PDMS. In aligned tissues, the anisotropy ratio of CPV (longitudinal CPV/transverse CPV) was lower on micropatterned soft PDMS compared to micropatterned stiff PDMS and micromolded gelatin hydrogels. Conclusion Propagation velocity in engineered cardiac tissues is sensitive to features in the tissue microenvironment, such as alignment, matrix elasticity, and matrix topography, which may underlie arrhythmias in conditions with pathological tissue remodeling.
Collapse
|
38
|
Pasqualini FS, Agarwal A, O'Connor BB, Liu Q, Sheehy SP, Parker KK. Traction force microscopy of engineered cardiac tissues. PLoS One 2018; 13:e0194706. [PMID: 29590169 PMCID: PMC5874032 DOI: 10.1371/journal.pone.0194706] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 03/08/2018] [Indexed: 01/08/2023] Open
Abstract
Cardiac tissue development and pathology have been shown to depend sensitively on microenvironmental mechanical factors, such as extracellular matrix stiffness, in both in vivo and in vitro systems. We present a novel quantitative approach to assess cardiac structure and function by extending the classical traction force microscopy technique to tissue-level preparations. Using this system, we investigated the relationship between contractile proficiency and metabolism in neonate rat ventricular myocytes (NRVM) cultured on gels with stiffness mimicking soft immature (1 kPa), normal healthy (13 kPa), and stiff diseased (90 kPa) cardiac microenvironments. We found that tissues engineered on the softest gels generated the least amount of stress and had the smallest work output. Conversely, cardiomyocytes in tissues engineered on healthy- and disease-mimicking gels generated significantly higher stresses, with the maximal contractile work measured in NRVM engineered on gels of normal stiffness. Interestingly, although tissues on soft gels exhibited poor stress generation and work production, their basal metabolic respiration rate was significantly more elevated than in other groups, suggesting a highly ineffective coupling between energy production and contractile work output. Our novel platform can thus be utilized to quantitatively assess the mechanotransduction pathways that initiate tissue-level structural and functional remodeling in response to substrate stiffness.
Collapse
Affiliation(s)
- Francesco Silvio Pasqualini
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, United States of America
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, United States of America
| | - Ashutosh Agarwal
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, United States of America
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, United States of America
- Department of Biomedical Engineering, University of Miami, Miami, FL, United States of America
- Department of Pathology, University of Miami Miller School of Medicine, Miami, FL, United States of America
- Dr. John T. Macdonald Foundation Biomedical Nanotechnology Institute, Miami, FL, United States of America
| | - Blakely Bussie O'Connor
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, United States of America
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, United States of America
| | - Qihan Liu
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, United States of America
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, United States of America
| | - Sean P. Sheehy
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, United States of America
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, United States of America
| | - Kevin Kit Parker
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, United States of America
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, United States of America
- * E-mail:
| |
Collapse
|
39
|
Cho N, Razipour SE, McCain ML. Featured Article: TGF-β1 dominates extracellular matrix rigidity for inducing differentiation of human cardiac fibroblasts to myofibroblasts. Exp Biol Med (Maywood) 2018; 243:601-612. [PMID: 29504479 DOI: 10.1177/1535370218761628] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cardiac fibroblasts and their activated derivatives, myofibroblasts, play a critical role in wound healing after myocardial injury and often contribute to long-term pathological outcomes, such as excessive fibrosis. Thus, defining the microenvironmental factors that regulate the phenotype of cardiac fibroblasts and myofibroblasts could lead to new therapeutic strategies. Both chemical and biomechanical cues have previously been shown to induce myofibroblast differentiation in many organs and species. For example, transforming growth factor beta 1, a cytokine secreted by neutrophils, and rigid extracellular matrix environments have both been shown to promote differentiation. However, the relative contributions of transforming growth factor beta 1 and extracellular matrix rigidity, two hallmark cues in many pathological myocardial microenvironments, to the phenotype of human cardiac fibroblasts are unclear. We hypothesized that transforming growth factor beta 1 and rigid extracellular matrix environments would potentially have a synergistic effect on the differentiation of human cardiac fibroblasts to myofibroblasts. To test this, we seeded primary human adult cardiac fibroblasts onto coverslips coated with polydimethylsiloxane of various elastic moduli, introduced transforming growth factor beta 1, and longitudinally quantified cell phenotype by measuring expression of α-smooth muscle actin, the most robust indicator of myofibroblasts. Our data indicate that, although extracellular matrix rigidity influenced differentiation after one day of transforming growth factor beta 1 treatment, ultimately transforming growth factor beta 1 superseded extracellular matrix rigidity as the primary regulator of myofibroblast differentiation. We also measured expression of POSTN, FAP, and FSP1, proposed secondary indicators of fibroblast/myofibroblast phenotypes. Although these genes partially trended with α-smooth muscle actin expression, they were relatively inconsistent. Finally, we demonstrated that activated myofibroblasts incompletely revert to a fibroblast phenotype after they are re-plated onto new surfaces without transforming growth factor beta 1, suggesting differentiation is partially reversible. Our results provide new insights into how microenvironmental cues affect human cardiac fibroblast differentiation in the context of myocardial pathology, which is important for identifying effective therapeutic targets and dictating supporting cell phenotypes for engineered human cardiac disease models. Impact statement Heart disease is the leading cause of death worldwide. Many forms of heart disease are associated with fibrosis, which increases extracellular matrix (ECM) rigidity and compromises cardiac output. Fibrotic tissue is synthesized primarily by myofibroblasts differentiated from fibroblasts. Thus, defining the cues that regulate myofibroblast differentiation is important for understanding the mechanisms of fibrosis. However, previous studies have focused on non-human cardiac fibroblasts and have not tested combinations of chemical and mechanical cues. We tested the effects of TGF-β1, a cytokine secreted by immune cells after injury, and ECM rigidity on the differentiation of human cardiac fibroblasts to myofibroblasts. Our results indicate that differentiation is initially influenced by ECM rigidity, but is ultimately superseded by TGF-β1. This suggests that targeting TGF-β signaling pathways in cardiac fibroblasts may have therapeutic potential for attenuating fibrosis, even in rigid microenvironments. Additionally, our approach can be leveraged to engineer more precise multi-cellular human cardiac tissue models.
Collapse
Affiliation(s)
- Nathan Cho
- 1 Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Shadi E Razipour
- 1 Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Megan L McCain
- 1 Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA.,2 Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
40
|
Ulmer BM, Stoehr A, Schulze ML, Patel S, Gucek M, Mannhardt I, Funcke S, Murphy E, Eschenhagen T, Hansen A. Contractile Work Contributes to Maturation of Energy Metabolism in hiPSC-Derived Cardiomyocytes. Stem Cell Reports 2018; 10:834-847. [PMID: 29503093 PMCID: PMC5919410 DOI: 10.1016/j.stemcr.2018.01.039] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 01/29/2018] [Accepted: 01/30/2018] [Indexed: 02/07/2023] Open
Abstract
Energy metabolism is a key aspect of cardiomyocyte biology. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are a promising tool for biomedical application, but they are immature and have not undergone metabolic maturation related to early postnatal development. To assess whether cultivation of hiPSC-CMs in 3D engineered heart tissue format leads to maturation of energy metabolism, we analyzed the mitochondrial and metabolic state of 3D hiPSC-CMs and compared it with 2D culture. 3D hiPSC-CMs showed increased mitochondrial mass, DNA content, and protein abundance (proteome). While hiPSC-CMs exhibited the principal ability to use glucose, lactate, and fatty acids as energy substrates irrespective of culture format, hiPSC-CMs in 3D performed more oxidation of glucose, lactate, and fatty acid and less anaerobic glycolysis. The increase in mitochondrial mass and DNA in 3D was diminished by pharmacological reduction of contractile force. In conclusion, contractile work contributes to metabolic maturation of hiPSC-CMs. Higher mitochondrial mass, protein, and DNA content in 3D versus 2D hiPSC-CMs Similarity of mitochondrial proteomes between 3D hiPSC-CMs and adult human heart Preference for oxidative metabolism in favor of anaerobic glycolysis in 3D hiPSC-CMs
Collapse
Affiliation(s)
- Bärbel M Ulmer
- University Medical Center Hamburg Eppendorf, Department of Experimental Pharmacology and Toxicology, 20246 Hamburg, Germany; German Center for Heart Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany.
| | - Andrea Stoehr
- Systems Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mirja L Schulze
- University Medical Center Hamburg Eppendorf, Department of Experimental Pharmacology and Toxicology, 20246 Hamburg, Germany; German Center for Heart Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Sajni Patel
- Proteomics Core Facility, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Marjan Gucek
- Proteomics Core Facility, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ingra Mannhardt
- University Medical Center Hamburg Eppendorf, Department of Experimental Pharmacology and Toxicology, 20246 Hamburg, Germany; German Center for Heart Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Sandra Funcke
- University Medical Center Hamburg Eppendorf, Department of Experimental Pharmacology and Toxicology, 20246 Hamburg, Germany; German Center for Heart Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Elizabeth Murphy
- Systems Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Thomas Eschenhagen
- University Medical Center Hamburg Eppendorf, Department of Experimental Pharmacology and Toxicology, 20246 Hamburg, Germany; German Center for Heart Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Arne Hansen
- University Medical Center Hamburg Eppendorf, Department of Experimental Pharmacology and Toxicology, 20246 Hamburg, Germany; German Center for Heart Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany.
| |
Collapse
|
41
|
Bartolák-Suki E, Imsirovic J, Nishibori Y, Krishnan R, Suki B. Regulation of Mitochondrial Structure and Dynamics by the Cytoskeleton and Mechanical Factors. Int J Mol Sci 2017; 18:E1812. [PMID: 28825689 PMCID: PMC5578198 DOI: 10.3390/ijms18081812] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 08/09/2017] [Accepted: 08/18/2017] [Indexed: 12/21/2022] Open
Abstract
Mitochondria supply cells with energy in the form of ATP, guide apoptosis, and contribute to calcium buffering and reactive oxygen species production. To support these diverse functions, mitochondria form an extensive network with smaller clusters that are able to move along microtubules aided by motor proteins. Mitochondria are also associated with the actin network, which is involved in cellular responses to various mechanical factors. In this review, we discuss mitochondrial structure and function in relation to the cytoskeleton and various mechanical factors influencing cell functions. We first summarize the morphological features of mitochondria with an emphasis on fission and fusion as well as how network properties govern function. We then review the relationship between the mitochondria and the cytoskeletal structures, including mechanical interactions. We also discuss how stretch and its dynamic pattern affect mitochondrial structure and function. Finally, we present preliminary data on how extracellular matrix stiffness influences mitochondrial morphology and ATP generation. We conclude by discussing the more general role that mitochondria may play in mechanobiology and how the mechanosensitivity of mitochondria may contribute to the development of several diseases and aging.
Collapse
Affiliation(s)
| | - Jasmin Imsirovic
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA.
| | - Yuichiro Nishibori
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA.
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan.
| | - Ramaswamy Krishnan
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Béla Suki
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA.
| |
Collapse
|