1
|
Heisser RH, Bawa M, Shah J, Bu A, Raman R. Soft Biological Actuators for Meter-Scale Homeostatic Biohybrid Robots. Chem Rev 2025; 125:3976-4007. [PMID: 40138615 DOI: 10.1021/acs.chemrev.4c00785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Skeletal muscle's elegant protein-based architecture powers motion throughout the animal kingdom, with its constituent actomyosin complexes driving intra- and extra-cellular motion. Classical motors and recently developed soft actuators cannot match the packing density and contractility of individual muscle fibers that scale to power the motion of ants and elephants alike. Accordingly, the interdisciplinary fields of robotics and tissue engineering have combined efforts to build living muscle actuators that can power a new class of robots to be more energy-efficient, dexterous, and safe than existing motor-powered and hydraulic paradigms. Doing so ethically and at scale─creating meter-scale tissue constructs from sustainable muscle progenitor cell lines─has inspired innovations in biomaterials and tissue culture methodology. We weave discussions of muscle cell biology, materials chemistry, tissue engineering, and biohybrid design to review the state of the art in soft actuator biofabrication. Looking forward, we outline a vision for meter-scale biohybrid robotic systems and tie discussions of recent progress to long-term research goals.
Collapse
Affiliation(s)
- Ronald H Heisser
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, Massachusetts 02139, United States of America
| | - Maheera Bawa
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, Massachusetts 02139, United States of America
| | - Jessica Shah
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, Massachusetts 02139, United States of America
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, 45 Carleton St., Cambridge, Massachusetts 02142, United States of America
| | - Angel Bu
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, Massachusetts 02139, United States of America
| | - Ritu Raman
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, Massachusetts 02139, United States of America
| |
Collapse
|
2
|
Faiella M, Botti G, Dalpiaz A, Gnudi L, Goyenvalle A, Pavan B, Perrone D, Bovolenta M, Marchesi E. In Vitro Studies to Evaluate the Intestinal Permeation of an Ursodeoxycholic Acid-Conjugated Oligonucleotide for Duchenne Muscular Dystrophy Treatment. Pharmaceutics 2024; 16:1023. [PMID: 39204368 PMCID: PMC11360444 DOI: 10.3390/pharmaceutics16081023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 07/24/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Delivery represents a major hurdle to the clinical advancement of oligonucleotide therapeutics for the treatment of disorders such as Duchenne muscular dystrophy (DMD). In this preliminary study, we explored the ability of 2'-O-methyl-phosphorothioate antisense oligonucleotides (ASOs) conjugated with lipophilic ursodeoxycholic acid (UDCA) to permeate across intestinal barriers in vitro by a co-culture system of non-contacting IEC-6 cells and DMD myotubes, either alone or encapsulated in exosomes. UDCA was used to enhance the lipophilicity and membrane permeability of ASOs, potentially improving oral bioavailability. Exosomes were employed due to their biocompatibility and ability to deliver therapeutic cargo across biological barriers. Exon skipping was evaluated in the DMD myotubes to reveal the targeting efficiency. Exosomes extracted from milk and wild-type myotubes loaded with 5'-UDC-3'Cy3-ASO and seeded directly on DMD myotubes appear able to fuse to myotubes and induce exon skipping, up to ~20%. Permeation studies using the co-culture system were performed with 5'-UDC-3'Cy3-ASO 51 alone or loaded in milk-derived exosomes. In this setting, only gymnotic delivery induced significant levels of exon skipping (almost 30%) implying a possible role of the intestinal cells in enhancing delivery of ASOs. These results warrant further investigations to elucidate the delivery of ASOs by gymnosis or exosomes.
Collapse
Affiliation(s)
- Marika Faiella
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (M.F.); (M.B.)
| | - Giada Botti
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy; (G.B.); (A.D.); (E.M.)
- Center for Translational Neurophysiology of Speech and Communication (CTNSC@UniFe), Italian Institute of Technology (IIT), 44121 Ferrara, Italy
| | - Alessandro Dalpiaz
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy; (G.B.); (A.D.); (E.M.)
| | - Lorenzo Gnudi
- Department of Environmental and Prevention Sciences, University of Ferrara, 44121 Ferrara, Italy;
| | - Aurélie Goyenvalle
- University Paris-Saclay, UVSQ, Inserm, END-ICAP, 78000 Versailles, France;
| | - Barbara Pavan
- Center for Translational Neurophysiology of Speech and Communication (CTNSC@UniFe), Italian Institute of Technology (IIT), 44121 Ferrara, Italy
- Department of Neuroscience and Rehabilitation—Section of Physiology, University of Ferrara, 44121 Ferrara, Italy
| | - Daniela Perrone
- Department of Environmental and Prevention Sciences, University of Ferrara, 44121 Ferrara, Italy;
| | - Matteo Bovolenta
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (M.F.); (M.B.)
| | - Elena Marchesi
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy; (G.B.); (A.D.); (E.M.)
| |
Collapse
|
3
|
Peng Z, Ahsan N, Yang Z. Proteomics Analysis of Interactions between Drug-Resistant and Drug-Sensitive Cancer Cells: Comparative Studies of Monoculture and Coculture Cell Systems. J Proteome Res 2024; 23:2608-2618. [PMID: 38907724 PMCID: PMC11425778 DOI: 10.1021/acs.jproteome.4c00338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/24/2024]
Abstract
Cell-cell interactions, which allow cells to communicate with each other through molecules in their microenvironment, are critical for the growth, health, and functions of cells. Previous studies show that drug-resistant cells can interact with drug-sensitive cells to elevate their drug resistance level, which is partially responsible for cancer recurrence. Studying protein targets and pathways involved in cell-cell communication provides essential information for fundamental cell biology studies and therapeutics of human diseases. In the current studies, we performed direct coculture and indirect coculture of drug-resistant and drug-sensitive cell lines, aiming to investigate intracellular proteins responsible for cell communication. Comparative studies were carried out using monoculture cells. Shotgun bottom-up proteomics results indicate that the P53 signaling pathway has a strong association with drug resistance mechanisms, and multiple TP53-related proteins were upregulated in both direct and indirect coculture systems. In addition, cell-cell communication pathways, including the phagosome and the HIF-signaling pathway, contribute to both direct and indirect coculture systems. Consequently, AK3 and H3-3A proteins were identified as potential targets for cell-cell interactions that are relevant to drug resistance mechanisms. We propose that the P53 signaling pathway, in which mitochondrial proteins play an important role, is responsible for inducing drug resistance through communication between drug-resistant and drug-sensitive cancer cells.
Collapse
Affiliation(s)
- Zongkai Peng
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Nagib Ahsan
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
- Mass Spectrometry, Proteomics and Metabolomics Core Facility, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Zhibo Yang
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| |
Collapse
|
4
|
Sood A, Singhmar R, Son Y, Jo CH, Choi S, Kumar A, Soo Han S. Tuning the efficacy of decellularized apple by coating with alginate/gelatin to behave as a bioscaffold for cultured meat production. Food Res Int 2024; 177:113907. [PMID: 38225146 DOI: 10.1016/j.foodres.2023.113907] [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: 10/16/2023] [Revised: 12/13/2023] [Accepted: 12/21/2023] [Indexed: 01/17/2024]
Abstract
Substantial efforts are underway to tackle the current challenges of sustainability and environmental impacts linked to orthodox animal agriculture. This had led to advancement in food innovation guiding the fabrication of edible scaffolds based cultured meat. This current research work aims to develop and validate a new approach in fabricating a 3D porous scaffold of decellularized apple coated with a polymer mixture of gelatin/alginate for cultivated meat production. The fabricated noncoated (A) and coated (CA) 3D scaffolds presented different ratios of pore sizes with the medium-sized pores (100-250 µm) being higher in the case of CA. The water absorption capacity of CA (∼64 %) was almost two folds compared to A (∼31 %) with delayed digestion in the presence of gastric simulated juice with or without pepsin. Both the scaffolds showed the capability to adhere and proliferate muscle satellite cells as single cell culture and muscle satellite along with NIH/3T3 fibroblast cells as co-culture. However, the CA scaffolds showed enhanced capability to adhere and proliferate the two cell lines on its surface compared to A. This work demonstrates an efficient way to fabricate decellularized plant scaffolds with high potential to be used in the production of cultured meat for the food industry.
Collapse
Affiliation(s)
- Ankur Sood
- School of Chemical Engineering, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, South Korea.
| | - Ritu Singhmar
- School of Chemical Engineering, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, South Korea
| | - Yumi Son
- School of Chemical Engineering, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, South Korea
| | - Chae-Hyun Jo
- Core Research Support Centre for Natural Products and Medical Materials, 280 Daehak-ro, Gyeongsan 38541, South Korea
| | - Soonmo Choi
- School of Chemical Engineering, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, South Korea; Research Institute of Cell Culture, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, South Korea
| | - Anuj Kumar
- School of Chemical Engineering, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, South Korea; School of Materials Science and Technology, Indian Institute of Technology (BHU), Varanasi 221005, India.
| | - Sung Soo Han
- School of Chemical Engineering, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, South Korea; Research Institute of Cell Culture, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, South Korea.
| |
Collapse
|
5
|
Song Y, Wei D, Raza SHA, Zhao Y, Jiang C, Song X, Wu H, Wang X, Luoreng Z, Ma Y. Research progress of intramuscular fat formation based on co-culture. Anim Biotechnol 2023; 34:3216-3236. [PMID: 36200856 DOI: 10.1080/10495398.2022.2127410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
Intramuscular fat (IMF) is closely related to the meat quality of livestock and poultry. As a new cell culture technique in vitro, cell co-culture has been gradually applied to the related research of IMF formation because it can simulate the changes of microenvironment in vivo during the process of IMF cell formation. In the co-culture model, in addition to studying the effects of skeletal muscle cells on the proliferation and differentiation of IMF, we can also consider the role of many secretion factors in the formation of IMF, thus making the cell research in vitro closer to the real level in vivo. This paper reviewed the generation and origin of IMF, summarized the existing co-culture methods and systems, and discussed the advantages and disadvantages of each method as well as the challenges faced in the establishment of the system, with emphasis on the current status of research on the formation of IMF for human and animal based on co-culture technology.
Collapse
Affiliation(s)
- Yaping Song
- School of Agriculture, Ningxia University, Ningxia Yin Chuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia University, Ningxia Yinchuan, China
| | - Dawei Wei
- School of Agriculture, Ningxia University, Ningxia Yin Chuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia University, Ningxia Yinchuan, China
| | | | - Yiang Zhao
- School of Agriculture, Ningxia University, Ningxia Yin Chuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia University, Ningxia Yinchuan, China
| | - Chao Jiang
- School of Agriculture, Ningxia University, Ningxia Yin Chuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia University, Ningxia Yinchuan, China
| | - Xiaoyu Song
- School of Agriculture, Ningxia University, Ningxia Yin Chuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia University, Ningxia Yinchuan, China
| | - Hao Wu
- School of Agriculture, Ningxia University, Ningxia Yin Chuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia University, Ningxia Yinchuan, China
| | - Xingping Wang
- School of Agriculture, Ningxia University, Ningxia Yin Chuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia University, Ningxia Yinchuan, China
| | - Zhuoma Luoreng
- School of Agriculture, Ningxia University, Ningxia Yin Chuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia University, Ningxia Yinchuan, China
| | - Yun Ma
- School of Agriculture, Ningxia University, Ningxia Yin Chuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia University, Ningxia Yinchuan, China
| |
Collapse
|
6
|
miR-34a Regulates Lipid Droplet Deposition in 3T3-L1 and C2C12 Cells by Targeting LEF1. Cells 2022; 12:cells12010167. [PMID: 36611960 PMCID: PMC9818453 DOI: 10.3390/cells12010167] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/21/2022] [Accepted: 12/27/2022] [Indexed: 01/03/2023] Open
Abstract
Intramuscular fat (IMF) content plays a key role in improving the flavor and palatability of pork. The IMF content varies between species, breeds, and individuals of the same breed. Hence, it is necessary to elucidate the mechanisms of IMF deposition to improve pork quality. Herein, the IMF content in the longissimus dorsi muscles of 29 Laiwu pigs was detected and divided into two groups, the H group (IMF > 12%) and the L group (IMF < 5%). RNA sequencing analysis showed 24 differentially expressed (DE) miRNA, and GO and KEGG analysis demonstrated that the DE miRNAs were significantly enriched in lipid metabolic process, lipid storage, Wnt, mTOR, and PPAR signaling pathways. miR-34a was found to be increased in the H group and 3T3-L1-derived adipocytes, while Lef1 was decreased. Luciferase reporter assays demonstrated that Lef1 was a potential target of miR-34a. Mechanism analysis revealed that miR-34a could increase lipid droplet deposition in 3T3-L1 and C2C12 cells by dampening the suppressive function of Lef1 on the transcription of adipogenic markers (i.e., Pparg, Cebpa, Fabp4, and Plin1). Moreover, overexpression of miR-34a could enhance the lipid deposition in the co-culture system of 3T3-L1 and C2C12 cells as well as in C2C12 cells cultured with conditioned medium from the progress of adipocyte differentiation. Taken together, our study indicated that miR-34a was an important positive modulator in the regulation of fatty metabolism and fat deposition by inhibiting the suppressive function of Lef1. These results might provide insight for the exploration of potential strategies to promote intramuscular fat deposition in livestock.
Collapse
|
7
|
Yue B, Wang H, Cai X, Wang J, Chai Z, Peng W, Shu S, Fu C, Zhong J. Adipose-Secreted Exosomes and Their Pathophysiologic Effects on Skeletal Muscle. Int J Mol Sci 2022; 23:12411. [PMID: 36293266 PMCID: PMC9604254 DOI: 10.3390/ijms232012411] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/07/2022] [Accepted: 10/14/2022] [Indexed: 04/30/2024] Open
Abstract
Due to its prominent secretory activity, adipose tissue (AT) is now considered a major player in the crosstalk between organs, especially with skeletal muscle. In which, exosomes are effective carriers for the intercellular material transfer of a wide range of molecules that can influence a series of physiological and pathological processes in recipient cells. Considering their underlying roles, the regulatory mechanisms of adipose-secreted exosomes and their cellular crosstalk with skeletal muscle have received great attention in the field. In this review, we describe what is currently known of adipose-secreted exosomes, as well as their applications in skeletal muscle pathophysiology.
Collapse
Affiliation(s)
- Binglin Yue
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu 610225, China
| | - Hui Wang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu 610225, China
| | - Xin Cai
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu 610225, China
| | - Jiabo Wang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu 610225, China
| | - Zhixin Chai
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu 610225, China
| | - Wei Peng
- Academy of Animal Science and Veterinary Medicine, Qinghai University, Xining 810016, China
| | - Shi Shu
- Academy of Animal Science and Veterinary Medicine, Qinghai University, Xining 810016, China
| | - Changqi Fu
- Academy of Animal Science and Veterinary Medicine, Qinghai University, Xining 810016, China
| | - Jincheng Zhong
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu 610225, China
| |
Collapse
|
8
|
Chen X, Peng Z, Yang Z. Metabolomics studies of cell-cell interactions using single cell mass spectrometry combined with fluorescence microscopy. Chem Sci 2022; 13:6687-6695. [PMID: 35756524 PMCID: PMC9172575 DOI: 10.1039/d2sc02298b] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 05/15/2022] [Indexed: 11/21/2022] Open
Abstract
Cell-cell interactions are critical for transmitting signals among cells and maintaining their normal functions from the single-cell level to tissues. In cancer studies, interactions between drug-resistant and drug-sensitive cells play an important role in the development of chemotherapy resistance of tumors. As metabolites directly reflect the cell status, metabolomics studies provide insight into cell-cell communication. Mass spectrometry (MS) is a powerful tool for metabolomics studies, and single cell MS (SCMS) analysis can provide unique information for understanding interactions among heterogeneous cells. In the current study, we utilized a direct co-culture system (with cell-cell contact) to study metabolomics of single cells affected by cell-cell interactions in their living status. A fluorescence microscope was utilized to distinguish these two types of cells for SCMS metabolomics studies using the Single-probe SCMS technique under ambient conditions. Our results show that through interactions with drug-resistant cells, drug-sensitive cancer cells acquired significantly increased drug resistance and exhibited drastically altered metabolites. Further investigation found that the increased drug resistance was associated with multiple metabolism regulations in drug-sensitive cells through co-culture such as the upregulation of sphingomyelins lipids and lactic acid and the downregulation of TCA cycle intermediates. The method allows for direct MS metabolomics studies of individual cells labeled with fluorescent proteins or dyes among heterogeneous populations.
Collapse
Affiliation(s)
- Xingxiu Chen
- Chemistry and Biochemistry Department, University of Oklahoma Norman Oklahoma 73072 USA
| | - Zongkai Peng
- Chemistry and Biochemistry Department, University of Oklahoma Norman Oklahoma 73072 USA
| | - Zhibo Yang
- Chemistry and Biochemistry Department, University of Oklahoma Norman Oklahoma 73072 USA
| |
Collapse
|
9
|
Goines S, Deng M, Glasscott MW, Leung JWC, Dick JE. Enhancing scanning electrochemical microscopy's potential to probe dynamic co-culture systems via hyperspectral assisted-imaging. Analyst 2022; 147:2396-2404. [PMID: 35579029 PMCID: PMC9287841 DOI: 10.1039/d2an00319h] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2023]
Abstract
Precise determination of boundaries in co-culture systems is difficult to achieve with scanning electrochemical microscopy alone. Thus, biological scanning electrochemical microscope platforms generally consist of a scanning electrochemical microscope positioner mounted on the stage of an inverted microscope for correlated electrochemical and optical imaging. Use of a fluorescence microscope allows for site-specific fluorescence labeling to obtain more clearly resolved spatial and electrochemical data. Here, we construct a unique hyperspectral assisted-biological scanning electrochemical microscope platform to widen the scope of biological imaging. Specifically, we incorporate a variable fluorescence bandpass source into a biological scanning electrochemical microscope platform for simultaneous optical, spectral, and electrochemical imaging. Not only does this platform serve as a cost-effective alternative to white light laser imaging, but additionally it provides multi-functional analysis of biological samples. Here, we demonstrate the efficacy of our platform to discern the electrochemical contribution of site-specific cells by optically and spectroscopically resolving boundaries as well as cell types within a complex biological system.
Collapse
Affiliation(s)
- Sondrica Goines
- Department of Chemistry, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Mingchu Deng
- Department of Chemistry, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Matthew W Glasscott
- Department of Chemistry, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Justin W C Leung
- Department of Radiation Oncology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Jeffrey E Dick
- Department of Chemistry, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
10
|
Vepkhvadze TF, Vorotnikov AV, Popov DV. Electrical Stimulation of Cultured Myotubes in vitro as a Model of Skeletal Muscle Activity: Current State and Future Prospects. BIOCHEMISTRY (MOSCOW) 2021; 86:597-610. [PMID: 33993862 DOI: 10.1134/s0006297921050084] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Skeletal muscles comprise more than a third of human body mass and critically contribute to regulation of body metabolism. Chronic inactivity reduces metabolic activity and functional capacity of muscles, leading to metabolic and other disorders, reduced life quality and duration. Cellular models based on progenitor cells isolated from human muscle biopsies and then differentiated into mature fibers in vitro can be used to solve a wide range of experimental tasks. The review discusses the aspects of myogenesis dynamics and regulation, which might be important in the development of an adequate cell model. The main function of skeletal muscle is contraction; therefore, electrical stimulation is important for both successful completion of myogenesis and in vitro modeling of major processes induced in the skeletal muscle by acute or regular physical exercise. The review analyzes the drawbacks of such cellular model and possibilities for its optimization, as well as the prospects for its further application to address fundamental aspects of muscle physiology and biochemistry and explore cellular and molecular mechanisms of metabolic diseases.
Collapse
Affiliation(s)
- Tatiana F Vepkhvadze
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, 123007, Russia
| | - Alexander V Vorotnikov
- National Medical Research Center of Cardiology, Ministry of Healthcare of the Russian Federation, Moscow, 121552, Russia
| | - Daniil V Popov
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, 123007, Russia. .,Faculty of Fundamental Medicine, Lomonosov Moscow State University, Moscow, 119991, Russia
| |
Collapse
|
11
|
Xu X, Zhao R, Ma W, Zhao Q, Zhang G. Comparison of lipid deposition of intramuscular preadipocytes in Tan sheep co-cultured with satellite cells or alone. J Anim Physiol Anim Nutr (Berl) 2021; 106:733-741. [PMID: 34189825 DOI: 10.1111/jpn.13599] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 06/07/2021] [Accepted: 06/08/2021] [Indexed: 01/21/2023]
Abstract
The purpose of this study was to investigate the effect of the skeletal muscle satellite cells (SMSCs) on the lipid deposition of the intramuscular preadipocytes (IMPs) in a co-culture system of the Tan sheep cells. The SMSCs and IMPs from Tan sheep were separated and cultured. After the two kinds of cells were separated and cultured, they were inoculated onto a transwell cell chamber co-culture plate for co-cultivation. When the cell density reached more than 90%, the cells were induced to differentiate. After the induction of the SMSCs differentiation for 8 days, the level of the IMPs differentiation and the expression levels of the differentiation marker genes and the key enzymes of the lipid metabolism were assessed. The results showed that the number and area of the lipid droplets in the IMPs in the co-culture system were significantly reduced compared to those in the IMPs culture alone (p < 0.05). Meanwhile, the expression levels of the PPARγ, c/EBPα, ACC, FAS mRNA in the IMPs were significantly decreased (p < 0.05); the expression level of aP2 mRNA was decreased, but the difference was not significant (p > 0.05).These findings indicate that the SMSCs of the Tan sheep in the co-culture system inhibited the lipid deposition by the IMPs.
Collapse
Affiliation(s)
- Xiaochun Xu
- North Minzu University/Collaborative Innovation Center for Food Production and Safety, Yinchuan, China
| | - Rui Zhao
- North Minzu University/Collaborative Innovation Center for Food Production and Safety, Yinchuan, China
| | - Wenping Ma
- North Minzu University/Collaborative Innovation Center for Food Production and Safety, Yinchuan, China
| | - Qingmei Zhao
- North Minzu University/Collaborative Innovation Center for Food Production and Safety, Yinchuan, China
| | - Guijie Zhang
- Ningxia University/School of Agriculture, Yinchuan, China
| |
Collapse
|
12
|
Acosta FM, Jia UTA, Stojkova K, Howland KK, Guda T, Pacelli S, Brey EM, Rathbone CR. Diabetic Conditions Confer Metabolic and Structural Modifications to Tissue-Engineered Skeletal Muscle. Tissue Eng Part A 2021; 27:549-560. [PMID: 32878567 PMCID: PMC8126424 DOI: 10.1089/ten.tea.2020.0138] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 08/06/2020] [Indexed: 12/12/2022] Open
Abstract
Skeletal muscle is a tissue that is directly involved in the progression and persistence of type 2 diabetes (T2D), a disease that is becoming increasingly common. Gaining better insight into the mechanisms that are affecting skeletal muscle dysfunction in the context of T2D has the potential to lead to novel treatments for a large number of patients. Through its ability to emulate skeletal muscle architecture while also incorporating aspects of disease, tissue-engineered skeletal muscle (TE-SkM) has the potential to provide a means for rapid high-throughput discovery of therapies to treat skeletal muscle dysfunction, to include that which occurs with T2D. Muscle precursor cells isolated from lean or obese male Zucker diabetic fatty rats were used to generate TE-SkM constructs. Some constructs were treated with adipogenic induction media to accentuate the presence of adipocytes that is a characteristic feature of T2D skeletal muscle. The maturity (compaction and creatine kinase activity), mechanical integrity (Young's modulus), organization (myotube orientation), and metabolic capacity (insulin-stimulated glucose uptake) were all reduced by diabetes. Treating constructs with adipogenic induction media increased the quantity of lipid within the diabetic TE-SkM constructs, and caused changes in construct compaction, cell orientation, and insulin-stimulated glucose uptake in both lean and diabetic samples. Collectively, the findings herein suggest that the recapitulation of structural and metabolic aspects of T2D can be accomplished by engineering skeletal muscle in vitro.
Collapse
Affiliation(s)
- Francisca M. Acosta
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, Texas, USA
- UTSA-UTHSCSA Joint Graduate Program in Biomedical Engineering, San Antonio, Texas, USA
| | - U-Ter Aonda Jia
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, Texas, USA
- UTSA-UTHSCSA Joint Graduate Program in Biomedical Engineering, San Antonio, Texas, USA
| | - Katerina Stojkova
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, Texas, USA
| | - Kennedy K. Howland
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, Texas, USA
| | - Teja Guda
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, Texas, USA
| | - Settimio Pacelli
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, Texas, USA
| | - Eric M. Brey
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, Texas, USA
| | - Christopher R. Rathbone
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, Texas, USA
| |
Collapse
|
13
|
Effect of Neudesin Neurotrophic Factor on Differentiation of Bovine Preadipocytes and Myoblasts in a Co-Culture System. Animals (Basel) 2020; 11:ani11010034. [PMID: 33375362 PMCID: PMC7823575 DOI: 10.3390/ani11010034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/17/2020] [Accepted: 12/23/2020] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Marbling beef refers to the red and white beef formed by depositing fat between muscle fibers, and “marbling” is an important factor affecting beef quality. Therefore, we established a co-culture system of adipocytes and myocytes to mimic the microenvironment of marbling beef in vivo. However, Neudesin neurotrophic factor (NENF) was not detected in the co-culture system, but was detected in both adipocytes and myoblasts cultured separately. Further studies revealed that NENF knockdown inhibits adipogenesis and promotes myogenesis in separately cultured preadipocytes and myoblasts. However, because the monoculture system does not include the interaction of bovine adipocytes and myoblasts in the formation of marbling beef, in this study we investigated the effect of recombinant protein NENF on the differentiation of adipocytes and myoblasts in the co-culture system. The addition of NENF inhibited the formation of lipid droplets in co-cultured adipocytes but had no significant effect on myotube formation. These results were different from, and even conflicted with, those in the monocultures, which suggested that regulation of NENF expression in the same cell type changed along with the cell microenvironment and the molecular mechanism of marbling beef formation cannot be fully revealed through studies on the monoculture system. Abstract In this study, we successfully established a co-culture system of bovine preadipocytes and myoblasts to explore the effect of exogenous addition of Neudesin neurotrophic factor (NENF) recombinant protein on the differentiation of adipocytes and myoblasts in co-culture. The optimal concentration of NENF recombinant protein was 100 pg/mL. NENF promoted the differentiation of bovine preadipocytes and inhibited the differentiation of bovine myoblasts when the cells were cultured separately. After adding NENF recombinant protein to the co-culture system, the accumulation of lipid droplets in bovine preadipocytes decreased, but the differentiation of bovine myoblasts did not change significantly. The results of real-time quantitative PCR (RT-qPCR) and Western blot showed that the expression levels of adipogenesis-related factors such as PPARγ, FABP4 and FASN were significantly down-regulated at the mRNA and protein levels in adipocytes, while myogenic marker genes such as MYOD1, MYOG and MYHC had no significant changes at the mRNA or protein levels in myoblasts. This differs from, and potentially conflicts with, the monoculture system, where NENF expression in each cell type changed with the cell microenvironment. Consequently, the molecular mechanism of marbling beef formation cannot be fully revealed using monocultures of adipocytes or myocytes.
Collapse
|
14
|
Adipose and Muscle Cell Co-Culture System: A Novel In Vitro Tool to Mimic the In Vivo Cellular Environment. BIOLOGY 2020; 10:biology10010006. [PMID: 33374127 PMCID: PMC7823969 DOI: 10.3390/biology10010006] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/14/2020] [Accepted: 12/23/2020] [Indexed: 12/18/2022]
Abstract
A co-culture system allows researchers to investigate the complex interactions between two cell types under various environments, such as those that promote differentiation and growth as well as those that mimic healthy and diseased states, in vitro. In this paper, we review the most common co-culture systems for myocytes and adipocytes. The in vitro techniques mimic the in vivo environment and are used to investigate the causal relationships between different cell lines. Here, we briefly discuss mono-culture and co-culture cell systems and their applicability to the study of communication between two or more cell types, including adipocytes and myocytes. Also, we provide details about the different types of co-culture systems and their applicability to the study of metabolic disease, drug development, and the role of secretory factors in cell signaling cascades. Therefore, this review provides details about the co-culture systems used to study the complex interactions between adipose and muscle cells in various environments, such as those that promote cell differentiation and growth and those used for drug development.
Collapse
|
15
|
Seo K, Suzuki T, Kobayashi K, Nishimura T. Adipocytes suppress differentiation of muscle cells in a co-culture system. Anim Sci J 2018; 90:423-434. [PMID: 30585366 DOI: 10.1111/asj.13145] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 10/16/2018] [Accepted: 11/02/2018] [Indexed: 12/18/2022]
Abstract
The development of adipose tissue in skeletal muscle is important for improving meat quality. However, it is still unclear how adipocytes grow in the proximity of muscle fibers. We hypothesized that adipocytes would suppress muscle cell growth so as to grow dominantly within muscle. In this study, we investigated the effect of adipocytes on the differentiation of muscle cells in a co-culture system. The fusion index of C2C12 myoblasts co-cultured with 3T3-L1 adipocytes was significantly lower than that of the control. The expression of myogenin and myosin heavy chain in C2C12 muscle cells co-cultured with 3T3-L1 adipocytes was significantly lower than in the control. Furthermore, the expression of Atrogin-1 and MuRF-1 was higher in C2C12 muscle cells co-cultured with 3T3-L1 adipocytes than the control. These results suggest that 3T3-L1 adipocytes suppress the differentiation of C2C12 myoblasts. In addition, 3T3-L1 adipocytes induced the expression and secretion of IL-6 in C2C12 muscle cells. The fusion index and myotube diameter were higher in C2C12 muscle cells co-cultured with 3T3-L1 cells in medium containing IL-6-neutralizing antibody than the control. Taken together, there is a possibility that adipocyte-induced IL-6 expression in muscle cells could be involved in the inhibition of muscle cell differentiation via autocrine.
Collapse
Affiliation(s)
- Kangmin Seo
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Graduate School of Agriculture, Hokkaido University, Sapporo, Japan
| | - Takahiro Suzuki
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Graduate School of Agriculture, Hokkaido University, Sapporo, Japan
| | - Ken Kobayashi
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Graduate School of Agriculture, Hokkaido University, Sapporo, Japan
| | - Takanori Nishimura
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Graduate School of Agriculture, Hokkaido University, Sapporo, Japan
| |
Collapse
|
16
|
Guo L, Cui H, Zhao G, Liu R, Li Q, Zheng M, Guo Y, Wen J. Intramuscular preadipocytes impede differentiation and promote lipid deposition of muscle satellite cells in chickens. BMC Genomics 2018; 19:838. [PMID: 30477424 PMCID: PMC6258484 DOI: 10.1186/s12864-018-5209-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 10/30/2018] [Indexed: 12/14/2022] Open
Abstract
Background Skeletal muscle satellite cells (MSC) are crucial for postnatal growth and regeneration of skeletal muscle. An interaction exists between MSC and intramuscular preadipocytes (IMPA). This study is the first to investigate the effects of IMPA on MSC in chickens and unveil the molecular mechanisms by transcriptome analysis. Results Primary MSC and IMPA were isolated from the pectoralis major muscle of 7-day-old chickens. After both cell types reached confluence, MSC were cultured alone or co-cultured with IMPA for 2 or 4 d. MSC treated for 2 d were subjected to RNA-seq. A total of 1653 known differentially expressed genes (DEG) were identified between co-cultured and mono-cultured MSC (|log2 FC| ≥ 1, FDR < 0.01). Based on Gene Ontology analysis, 48 DEG related to muscle development were screened, including the key genes MYOD1, MYOG, PAX7, and TMEM8C. The 44 DEG related to lipid deposition included the key genes CD36, FABP4, ACSBG2, CYP7A1 and PLIN2. Most of the DEG related to muscle development were downregulated in co-cultured MSC, and DEG related to lipid deposition were upregulated. Immunofluorescence of MHC supported IMPA impeding differentiation of MSC, and Oil Red O staining showed concurrent promotion of lipid deposition. Pathway analysis found that several key genes were enriched in JNK/MAPK and PPAR signaling, which may be the key pathways regulating differentiation and lipid deposition in MSC. Additionally, pathways related to cell junctions may also contribute to the effect of IMPA on MSC. Conclusions The present study showed that IMPA impeded differentiation of MSC while promoting their lipid deposition. Pathway analysis indicated that IMPA might inhibit differentiation via the JNK/MAPK pathway, and promote lipid deposition via the PPAR pathway. This study supplies insights into the effect of IMPA on MSC, providing new clues on exposing the molecular mechanisms underlying the interplay between skeletal muscle and intramuscular fat in chickens. Electronic supplementary material The online version of this article (10.1186/s12864-018-5209-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Liping Guo
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.,College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.,State Key Laboratory of Animal Nutrition, Beijing, 100193, China
| | - Huanxian Cui
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.,State Key Laboratory of Animal Nutrition, Beijing, 100193, China
| | - Guiping Zhao
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.,State Key Laboratory of Animal Nutrition, Beijing, 100193, China
| | - Ranran Liu
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.,State Key Laboratory of Animal Nutrition, Beijing, 100193, China
| | - Qinghe Li
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.,State Key Laboratory of Animal Nutrition, Beijing, 100193, China
| | - Maiqing Zheng
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.,State Key Laboratory of Animal Nutrition, Beijing, 100193, China
| | - Yuming Guo
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China. .,State Key Laboratory of Animal Nutrition, Beijing, 100193, China.
| | - Jie Wen
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China. .,State Key Laboratory of Animal Nutrition, Beijing, 100193, China.
| |
Collapse
|
17
|
Li X, Tan R, Hu X, Jiao Q, Rahman MS, Chen X, Zhang P, An J, Lu H, Liu Y. Neural stem cell-derived factors inhibit the growth and invasion of U87 stem-like cells in vitro. J Cell Biochem 2018; 120:5472-5479. [PMID: 30367517 DOI: 10.1002/jcb.27826] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 09/12/2018] [Indexed: 01/05/2023]
Abstract
Glioma is one of the most common and aggressive tumors in the brain. Significant attention has been paid to the potential use of neural stem/progenitor cells (NSCs/NPCs) as delivery vehicles to cure gliomas. However, whether the NSCs/NPCs or the factors they produced could make a contribution still remains to be seen. In this study, we focused on the inhibitory effects of the factors produced by NSCs/NPCs on the biological behavior of the glioma stem-like cell in vitro. The human glioma cell line U87 was selected and the U87 stem-like cells were addressed. After being cultured in the NSC condition medium (NSC-CM), the viability and proliferation of U87 stem-like cells were significantly reduced. The invasion of U87 stem-like cells and the migration of U87 cells were also significantly decreased. However, no significant change was observed in regard to the astrocytic differentiation of U87 stem-like cells. These indicated that NSCs/NPCs produced some factors and had an inhibitory effect on the growth and invasion but not the terminal differentiation of U87 stem-like cells. It is worth paying attention to NSCs/NPCs as a high-potential candidate for glioma treatment.
Collapse
Affiliation(s)
- Xingxing Li
- Institute of Neurobiology, School of Basic Medical Sciences and Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Ruolan Tan
- Institute of Neurobiology, School of Basic Medical Sciences and Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Xiaoxuan Hu
- Institute of Neurobiology, School of Basic Medical Sciences and Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Qian Jiao
- Department of Physiology, Medical College of Qingdao University, Qingdao, China
| | - Md Saidur Rahman
- Institute of Neurobiology, School of Basic Medical Sciences and Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Xinlin Chen
- Institute of Neurobiology, School of Basic Medical Sciences and Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Pengbo Zhang
- Department of Anesthesiology, The Second Affiliated Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Jing An
- Institute of Neurobiology, School of Basic Medical Sciences and Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Haixia Lu
- Institute of Neurobiology, School of Basic Medical Sciences and Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Yong Liu
- Institute of Neurobiology, School of Basic Medical Sciences and Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, Xi'an, China
| |
Collapse
|
18
|
Lynes MD, Tseng YH. Deciphering adipose tissue heterogeneity. Ann N Y Acad Sci 2018; 1411:5-20. [PMID: 28763833 PMCID: PMC5788721 DOI: 10.1111/nyas.13398] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 05/02/2017] [Accepted: 05/05/2017] [Indexed: 02/06/2023]
Abstract
Obesity is an excess accumulation of adipose tissue mass, and, together with its sequelae, in particular type II diabetes and metabolic syndrome, obesity presents a major health crisis. Although obesity is simply caused by increased adipose mass, the heterogeneity of adipose tissue in humans means that the response to increased energy balance is highly complex. Individual subjects with similar phenotypes may respond very differently to the same treatments; therefore, obesity may benefit from a personalized precision medicine approach. The variability in the development of obesity is indeed driven by differences in sex, genetics, and environment, but also by the various types of adipose tissue as well as the different cell types that compose it. By describing the distinct cell populations that reside in different fat depots, we can interpret the complex effect of these various players in the maintenance of whole-body energy homeostasis. To further understand adipose tissue, adipogenic differentiation and the transcriptional program of lipid accumulation must be investigated. As the cell- and depot-specific functions are described, they can be placed in the context of energy excess to understand how the heterogeneity of adipose tissue shapes individual metabolic status and condition.
Collapse
Affiliation(s)
- Matthew D Lynes
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts and Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts
| | - Yu-Hua Tseng
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts and Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts
| |
Collapse
|
19
|
The inhibiting effect of neural stem cells on proliferation and invasion of glioma cells. Oncotarget 2017; 8:76949-76960. [PMID: 29100360 PMCID: PMC5652754 DOI: 10.18632/oncotarget.20270] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Accepted: 06/02/2017] [Indexed: 12/11/2022] Open
Abstract
The invasive and infiltrative nature of tumor cells leads to the poor prognosis of glioma. Currently, novel therapeutic means to eliminate the tumor cells without damaging the normal brain tissue are still strongly demanded. Significant attentions had been paid to stem cell-based therapy and neural stem cell (NSC) had been considered as one of the efficient delivery vehicles for targeting therapeutic genes. However, whether the NSCs could directly affect glioma cells remains to be seen. In this study, both rat and human glioma cells (C6 and U251) were co-cultured with normal rat embryonic NSCs directly or in-directly. We found the survival, proliferation, invasion and migration of glioma cells were significantly inhibited, while the differentiation was not affected in the in vitro co-culture system. In nude mice, although no significant difference was observed in the tumor growth, survival status and time of tumor-bearing mice were significantly promoted when U251 cells were subcutaneously injected with NSCs. In coincidence with the suppression of glioma cell growth in vitro, expression of mutant p53 and phosphorylation of AKT, ERK1/2 decreased while the level of caspase-3 increased significantly. Our results suggested that normal NSCs could possess direct anti-glioma properties via inhibiting the glioma cell viability, proliferation, invasion and migration. It could be a very promising candidate for glioma treatment.
Collapse
|
20
|
Pandurangan M, Mistry B, Enkhataivan G, Kim DH. Efficacy of carnosine on activation of caspase 3 and human renal carcinoma cell inhibition. Int J Biol Macromol 2016; 92:377-382. [DOI: 10.1016/j.ijbiomac.2016.07.044] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 07/11/2016] [Accepted: 07/13/2016] [Indexed: 11/28/2022]
|
21
|
Chandrasekaran M, Pandurangan M. In Vitro Selective Anti-Proliferative Effect of Zinc Oxide Nanoparticles Against Co-Cultured C2C12 Myoblastoma Cancer and 3T3-L1 Normal Cells. Biol Trace Elem Res 2016; 172:148-154. [PMID: 26563419 DOI: 10.1007/s12011-015-0562-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 10/29/2015] [Indexed: 02/07/2023]
Abstract
The zinc oxide (ZnO) nanoparticle has been widely used in biomedical applications and cancer therapy and has been reported to induce a selective cytotoxic effect on cancer cell proliferation. The present study investigated the cytotoxicity of ZnO nanoparticles against co-cultured C2C12 myoblastoma cancer cells and 3T3-L1 adipocytes. Our results showed that the ZnO nanoparticles could be cytotoxic to C2C12 myoblastoma cancer cells than 3T3-L1 cells. The messenger RNA (mRNA) expressions of p53 and bax were significantly increased 114.3 and 118.2 % in the C2C12 cells, whereas 42.5 and 40 % were increased in 3T3-L1 cells, respectively. The mRNA expression of bcl-2 was reduced 38.2 and 28.5 % in the C2C12 and 3T3-L1 cells, respectively, whereas the mRNA expression of caspase-3 was increased 80.7 and 51.6 % in the C2C12 and 3T3-L1 cells, respectively. The protein expressions of p53, bax, and caspase-3 were significantly increased 40, 81.8, and 80 % in C2C12 cells, whereas 20.3, 28.2, and 37.9 % were increased in 3T3-L1 cells, respectively. The mRNA expression of bcl-2 was significantly reduced 32.2 and 22.7 % in C2C12 and 3T3-L1 cells, respectively. Caspase-3 enzyme activity and reactive oxygen species (ROS) were increased in co-cultured C2C12 cells compared to 3T3-L1 cells. Taking all these data together, it may suggest that ZnO nanoparticles severely induce apoptosis in C2C12 myoblastoma cancer cells than 3T3-L1 cells.
Collapse
|
22
|
Chu W, Wei W, Yu S, Han H, Shi X, Sun W, Gao Y, Zhang L, Chen J. C2C12 myotubes inhibit the proliferation and differentiation of 3T3-L1 preadipocytes by reducing the expression of glucocorticoid receptor gene. Biochem Biophys Res Commun 2016; 472:68-74. [PMID: 26896766 DOI: 10.1016/j.bbrc.2016.02.063] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 02/15/2016] [Indexed: 12/15/2022]
Abstract
Obesity is a well-established risk factor to health for its relationship with insulin resistance, diabetes and metabolic syndrome. Myocyte-adipocyte crosstalk model plays a significant role in studying the interaction of muscle and adipose development. Previous related studies mainly focus on the effects of adipocytes on the myocytes activity, however, the influence of myotubes on the preadipocytes development remains unclear. The present study was carried out to settle this issue. Firstly, the co-culture experiment showed that the proliferation, cell cycle, and differentiation of 3T3-L1 preadipocytes were arrested, and the apoptosis was induced, by differentiated C2C12 myotubes. Next, the sensitivity of 3T3-L1 preadipocytes to glucocorticoids (GCs), which was well known as cell proliferation, differentiation, apoptosis factor, was decreased after co-cultured with C2C12 myotubes. What's more, our results showed that C2C12 myotubes suppressed the mRNA and protein expression of glucocorticoid receptor (GR) in 3T3-L1 preadipocytes, indicating the potential mechanism of GCs sensitivity reduction. Taken together, we conclude that C2C12 myotubes inhibited 3T3-L1 preadipocytes proliferation and differentiation by reducing the expression of GR. These data suggest that decreasing GR by administration of myokines may be a promising therapy for treating patients with obesity or diabetes.
Collapse
Affiliation(s)
- Weiwei Chu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Wei Wei
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Shigang Yu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Haiyin Han
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Xiaoli Shi
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Wenxing Sun
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, PR China; College of Public Health, Nantong University, Nantong 226019, PR China
| | - Ying Gao
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Lifan Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Jie Chen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, PR China.
| |
Collapse
|
23
|
Gizak A, Rakus D. Will Quantitative Proteomics Redefine Some of the Key Concepts in Skeletal Muscle Physiology? Proteomes 2016; 4:proteomes4010002. [PMID: 28248211 PMCID: PMC5217361 DOI: 10.3390/proteomes4010002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 12/21/2015] [Accepted: 01/05/2016] [Indexed: 01/28/2023] Open
Abstract
Molecular and cellular biology methodology is traditionally based on the reasoning called “the mechanistic explanation”. In practice, this means identifying and selecting correlations between biological processes which result from our manipulation of a biological system. In theory, a successful application of this approach requires precise knowledge about all parameters of a studied system. However, in practice, due to the systems’ complexity, this requirement is rarely, if ever, accomplished. Typically, it is limited to a quantitative or semi-quantitative measurements of selected parameters (e.g., concentrations of some metabolites), and a qualitative or semi-quantitative description of expression/post-translational modifications changes within selected proteins. A quantitative proteomics approach gives a possibility of quantitative characterization of the entire proteome of a biological system, in the context of the titer of proteins as well as their post-translational modifications. This enables not only more accurate testing of novel hypotheses but also provides tools that can be used to verify some of the most fundamental dogmas of modern biology. In this short review, we discuss some of the consequences of using quantitative proteomics to verify several key concepts in skeletal muscle physiology.
Collapse
Affiliation(s)
- Agnieszka Gizak
- Department of Animal Molecular Physiology, Wroclaw University, Cybulskiego 30, 50-205 Wroclaw, Poland.
| | - Dariusz Rakus
- Department of Animal Molecular Physiology, Wroclaw University, Cybulskiego 30, 50-205 Wroclaw, Poland.
| |
Collapse
|
24
|
Pandurangan M, Kim DH. A novel approach for in vitro meat production. Appl Microbiol Biotechnol 2015; 99:5391-5. [PMID: 25971200 DOI: 10.1007/s00253-015-6671-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2015] [Revised: 04/30/2015] [Accepted: 05/02/2015] [Indexed: 10/23/2022]
Abstract
The present review describes the possibility of in vitro meat production with the help of advanced co-culturing methods. In vitro meat production method could be a possible alternative for the conventional meat production. Originally, the research on in vitro meat production was initiated by the National Aeronautics and Space Administration (NASA) for space voyages. The required key qualities for accepting in vitro meat for consumption would be good efficiency ratio, increased protein synthesis rate in skeletal muscles, and mimicking the conventional meat qualities. In vitro culturing of meat is possible with the use of skeletal muscle tissue engineering, stem cell, cell co-culture, and tissue culture methods. Co-culture of myoblast and fibroblast is believed as one of the major techniques for in vitro meat production. In our lab, we have co-cultured myoblast and fibroblast. We believe that a billion pounds of in vitro meat could be produced from one animal for consumption. However, we require a great deal of research on in vitro meat production.
Collapse
|
25
|
Jin X, Yu H, Kong N, Chang J, Li H, Ye J. Superparamagnetic plasmonic nanoshells for improved imaging, separation and seeding of co-cultured cells. J Mater Chem B 2015; 3:7787-7795. [DOI: 10.1039/c5tb01420d] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Multifunctional superparamagnetic nanoshells were applied for improved 2D and 3D two-photon luminescence imaging, separation and seeding of co-cultured cells.
Collapse
Affiliation(s)
- Xiulong Jin
- School of Biomedical Engineering & Med-X Research Institute
- Shanghai Jiao Tong University
- Shanghai 200030
- China
| | - Hongfei Yu
- School of Biomedical Engineering & Med-X Research Institute
- Shanghai Jiao Tong University
- Shanghai 200030
- China
| | - Ni Kong
- School of Biomedical Engineering & Med-X Research Institute
- Shanghai Jiao Tong University
- Shanghai 200030
- China
| | - Jiang Chang
- Shanghai institute of Ceramics
- Chinese Academy of Sciences
- Shanghai 200050
- China
| | - Haiyan Li
- School of Biomedical Engineering & Med-X Research Institute
- Shanghai Jiao Tong University
- Shanghai 200030
- China
| | - Jian Ye
- School of Biomedical Engineering & Med-X Research Institute
- Shanghai Jiao Tong University
- Shanghai 200030
- China
| |
Collapse
|