51
|
Moretti B, Rodriguez Alvarez SN, Grecco HE. Nfinder: automatic inference of cell neighborhood in 2D and 3D using nuclear markers. BMC Bioinformatics 2023; 24:230. [PMID: 37270479 DOI: 10.1186/s12859-023-05284-2] [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: 03/29/2022] [Accepted: 04/12/2023] [Indexed: 06/05/2023] Open
Abstract
BACKGROUND In tissues and organisms, the coordination of neighboring cells is essential to maintain their properties and functions. Therefore, knowing which cells are adjacent is crucial to understand biological processes that involve physical interactions among them, e.g. cell migration and proliferation. In addition, some signaling pathways, such as Notch or extrinsic apoptosis, are highly dependent on cell-cell communication. While this is straightforward to obtain from membrane images, nuclei labelling is much more ubiquitous for technical reasons. However, there are no automatic and robust methods to find neighboring cells based only on nuclear markers. RESULTS In this work, we describe Nfinder, a method to assess the cell's local neighborhood from images with nuclei labeling. To achieve this goal, we approximate the cell-cell interaction graph by the Delaunay triangulation of nuclei centroids. Then, links are filtered by automatic thresholding in cell-cell distance (pairwise interaction) and the maximum angle that a pair of cells subtends with shared neighbors (non-pairwise interaction). We systematically characterized the detection performance by applying Nfinder to publicly available datasets from Drosophila melanogaster, Tribolium castaneum, Arabidopsis thaliana and C. elegans. In each case, the result of the algorithm was compared to a cell neighbor graph generated by manually annotating the original dataset. On average, our method detected 95% of true neighbors, with only 6% of false discoveries. Remarkably, our findings indicate that taking into account non-pairwise interactions might increase the Positive Predictive Value up to + 11.5%. CONCLUSION Nfinder is the first robust and automatic method for estimating neighboring cells in 2D and 3D based only on nuclear markers and without any free parameters. Using this tool, we found that taking non-pairwise interactions into account improves the detection performance significantly. We believe that using our method might improve the effectiveness of other workflows to study cell-cell interactions from microscopy images. Finally, we also provide a reference implementation in Python and an easy-to-use napari plugin.
Collapse
Affiliation(s)
- Bruno Moretti
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Física, Buenos Aires, Argentina.
- CONICET - Universidad de Buenos Aires, Instituto de Física de Buenos Aires (IFIBA), Buenos Aires, Argentina.
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, USA.
- Chan Zuckerberg Biohub-San Francisco, San Francisco, USA.
| | - Santiago N Rodriguez Alvarez
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Física, Buenos Aires, Argentina
- Institute of Physics, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Hernán E Grecco
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Física, Buenos Aires, Argentina.
- CONICET - Universidad de Buenos Aires, Instituto de Física de Buenos Aires (IFIBA), Buenos Aires, Argentina.
| |
Collapse
|
52
|
Barry-Carroll L, Greulich P, Marshall AR, Riecken K, Fehse B, Askew KE, Li K, Garaschuk O, Menassa DA, Gomez-Nicola D. Microglia colonize the developing brain by clonal expansion of highly proliferative progenitors, following allometric scaling. Cell Rep 2023; 42:112425. [PMID: 37099424 DOI: 10.1016/j.celrep.2023.112425] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 01/25/2023] [Accepted: 04/06/2023] [Indexed: 04/27/2023] Open
Abstract
Microglia arise from the yolk sac and enter the brain during early embryogenesis. Upon entry, microglia undergo in situ proliferation and eventually colonize the entire brain by the third postnatal week in mice. However, the intricacies of their developmental expansion remain unclear. Here, we characterize the proliferative dynamics of microglia during embryonic and postnatal development using complementary fate-mapping techniques. We demonstrate that the developmental colonization of the brain is facilitated by clonal expansion of highly proliferative microglial progenitors that occupy spatial niches throughout the brain. Moreover, the spatial distribution of microglia switches from a clustered to a random pattern between embryonic and late postnatal development. Interestingly, the developmental increase in microglial numbers follows the proportional growth of the brain in an allometric manner until a mosaic distribution has been established. Overall, our findings offer insight into how the competition for space may drive microglial colonization by clonal expansion during development.
Collapse
Affiliation(s)
- Liam Barry-Carroll
- School of Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Philip Greulich
- School of Mathematical Sciences, University of Southampton, Southampton, UK; Institute for Life Sciences (IfLS), University of Southampton, Southampton, UK
| | - Abigail R Marshall
- School of Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Kristoffer Riecken
- Research Department Cell and Gene Therapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Boris Fehse
- Research Department Cell and Gene Therapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katharine E Askew
- School of Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Kaizhen Li
- Department of Neurophysiology, University of Tübingen, Tübingen, Germany
| | - Olga Garaschuk
- Department of Neurophysiology, University of Tübingen, Tübingen, Germany
| | - David A Menassa
- School of Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK; The Queen's College, University of Oxford, Oxford, UK
| | - Diego Gomez-Nicola
- School of Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK; Institute for Life Sciences (IfLS), University of Southampton, Southampton, UK.
| |
Collapse
|
53
|
Ebata H, Umeda K, Nishizawa K, Nagao W, Inokuchi S, Sugino Y, Miyamoto T, Mizuno D. Activity-dependent glassy cell mechanics Ⅰ: Mechanical properties measured with active microrheology. Biophys J 2023; 122:1781-1793. [PMID: 37050875 PMCID: PMC10209042 DOI: 10.1016/j.bpj.2023.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 01/27/2023] [Accepted: 04/07/2023] [Indexed: 04/14/2023] Open
Abstract
Active microrheology was conducted in living cells by applying an optical-trapping force to vigorously fluctuating tracer beads with feedback-tracking technology. The complex shear modulus G(ω)=G'(ω)-iG″(ω) was measured in HeLa cells in an epithelial-like confluent monolayer. We found that G(ω)∝(-iω)1/2 over a wide range of frequencies (1 Hz < ω/2π < 10 kHz). Actin disruption and cell-cycle progression from G1 to S and G2 phases only had a limited effect on G(ω) in living cells. On the other hand, G(ω) was found to be dependent on cell metabolism; ATP-depleted cells showed an increased elastic modulus G'(ω) at low frequencies, giving rise to a constant plateau such that G(ω)=G0+A(-iω)1/2. Both the plateau and the additional frequency dependency ∝(-iω)1/2 of ATP-depleted cells are consistent with a rheological response typical of colloidal jamming. On the other hand, the plateau G0 disappeared in ordinary metabolically active cells, implying that living cells fluidize their internal states such that they approach the critical jamming point.
Collapse
Affiliation(s)
- Hiroyuki Ebata
- Department of Physics, Kyushu University, Fukuoka, Japan
| | | | - Kenji Nishizawa
- Institute of Developmental Biology of Marseille, Marseille, France
| | - Wataru Nagao
- Department of Physics, Kyushu University, Fukuoka, Japan
| | - Shono Inokuchi
- Department of Physics, Kyushu University, Fukuoka, Japan
| | - Yujiro Sugino
- Department of Physics, Kyushu University, Fukuoka, Japan
| | - Takafumi Miyamoto
- Department of Endocrinology and Metabolism, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan; Transborder Medical Research Center, University of Tsukuba, Ibaraki, Japan
| | - Daisuke Mizuno
- Department of Physics, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
54
|
Ning Y, Zheng H, Yang Y, Zang H, Wang W, Zhan Y, Wang H, Luo J, Wen Q, Peng J, Xiang J, Fan S. YAP1 synergize with YY1 transcriptional co-repress DUSP1 to induce osimertinib resistant by activating the EGFR/MAPK pathway and abrogating autophagy in non-small cell lung cancer. Int J Biol Sci 2023; 19:2458-2474. [PMID: 37215986 PMCID: PMC10197898 DOI: 10.7150/ijbs.79965] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 04/19/2023] [Indexed: 05/24/2023] Open
Abstract
YAP1 is a well-known core effector of the Hippo pathway in tumors, but its potential role in osimertinib resistance remained unexplored. Our study provides evidence that YAP1 acts as a potent promoter of osimertinib resistance. By inhibiting YAP1 with a novel inhibitor, CA3, and combining it with osimertinib, we observed a significant suppression of cell proliferation and metastasis, induction of apoptosis and autophagy, and a delay in the emergence of osimertinib resistance. Interestingly, CA3 combined with osimertinib executed its anti-metastasis and pro-tumor apoptosis in part through autophagy. Mechanistically, we found that YAP1, in collaboration with YY1, transcriptionally represses DUSP1, leading to the dephosphorylation of the EGFR/MEK/ERK pathway and YAP1 phosphorylation in osimertinib-resistant cells. Our results also validate that CA3, in combination with osimertinib, executes its anti-metastasis and pro-tumor apoptosis partly through autophagy and the YAP1/DUSP1/EGFR/MEK/ERK regulatory feedback loop in osimertinib-resistant cells. Remarkably, our findings illustrate that YAP1 protein is upregulated in patients after osimertinib treatment and osimertinib resistance. Overall, our study confirms that the YAP1 inhibitor CA3 increases DUSP1 with concomitant activation of the EGFR/MAPK pathway and induces autophagy to enhance the efficacy of third-generation EGFR-TKI treatments for NSCLC patients.
Collapse
Affiliation(s)
- Yue Ning
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hongmei Zheng
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yang Yang
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hongjing Zang
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Weiyuan Wang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuting Zhan
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Haihua Wang
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiadi Luo
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qiuyuan Wen
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jinwu Peng
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Juanjuan Xiang
- Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Songqing Fan
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
55
|
Jian Z, Li Y, Zhang C, Zhong W, Ai D, He Y, Song J. Low-Intensity Pulsed Ultrasound Attenuates Periodontal Ligament Cells Apoptosis by Activating Yes-Associated Protein-Regulated Autophagy. ULTRASOUND IN MEDICINE & BIOLOGY 2023; 49:1227-1237. [PMID: 36878833 DOI: 10.1016/j.ultrasmedbio.2023.01.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/24/2022] [Accepted: 01/11/2023] [Indexed: 05/11/2023]
Abstract
OBJECTIVE The goal of the work described here was to determine if low-intensity pulsed ultrasound (LIPUS) has an anti-inflammatory effect on lipopolysaccharide (LPS)-induced inflammation in periodontal ligament cells (PDLCs). The mechanism underlying this effect remains to be explored and is likely related to PDLC apoptosis regulated by Yes-associated protein (YAP) and autophagy. METHODS To verify this hypothesis, we used a rat model of periodontitis and primary human PDLCs. We examined alveolar bone resorption in rats and apoptosis, autophagy and YAP activity in LPS-treated PDLCs with and without application of LIPUS by cellular immunofluorescence, transmission electron microscopy and Western blotting. Then, siRNA transfection was used to decrease YAP expression to confirm the regulatory role of YAP in the anti-apoptotic effect of LIPUS on PDLCs. DISCUSSION We found that LIPUS attenuated alveolar bone resorption in rats and this was accompanied by YAP activation. LIPUS inhibited hPDLC apoptosis by YAP activation, and promoted autophagic degradation to help autophagy completion. These effects were reversed after YAP expression was blocked. CONCLUSION LIPUS attenuates PDLC apoptosis by activating Yes-associated protein-regulated autophagy.
Collapse
Affiliation(s)
- Zixiang Jian
- College of Stomatology, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Yao Li
- NMPA Key Laboratory for Dental Materials, National Engineering Laboratory for Digital and Material Technology of Stomatology, Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Peking, China
| | - Chuangwei Zhang
- College of Stomatology, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Wenjie Zhong
- College of Stomatology, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Dongqing Ai
- College of Stomatology, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Yao He
- College of Stomatology, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China.
| | - Jinlin Song
- College of Stomatology, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China.
| |
Collapse
|
56
|
Assi M, Kimmelman AC. Impact of context-dependent autophagy states on tumor progression. NATURE CANCER 2023; 4:596-607. [PMID: 37069394 PMCID: PMC10542907 DOI: 10.1038/s43018-023-00546-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 03/20/2023] [Indexed: 04/19/2023]
Abstract
Macroautophagy is a cellular quality-control process that degrades proteins, protein aggregates and damaged organelles. Autophagy plays a fundamental role in cancer where, in the presence of stressors (for example, nutrient starvation, hypoxia, mechanical pressure), tumor cells activate it to degrade intracellular substrates and provide energy. Cell-autonomous autophagy in tumor cells and cell-nonautonomous autophagy in the tumor microenvironment and in the host converge on mechanisms that modulate metabolic fitness, DNA integrity and immune escape and, consequently, support tumor growth. In this Review, we will discuss insights into the tumor-modulating roles of autophagy in different contexts and reflect on how future studies using physiological culture systems may help to understand the complexity and open new therapeutic avenues.
Collapse
Affiliation(s)
- Mohamad Assi
- Department of Radiation Oncology, New York University Langone Health, New York, NY, USA
- Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
| | - Alec C Kimmelman
- Department of Radiation Oncology, New York University Langone Health, New York, NY, USA.
- Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA.
| |
Collapse
|
57
|
Sathuvan M, Thangam R, Cheong KL, Kang H, Liu Y. κ-Carrageenan-essential oil loaded composite biomaterial film facilitates mechanosensing and tissue regenerative wound healing. Int J Biol Macromol 2023; 241:124490. [PMID: 37076080 DOI: 10.1016/j.ijbiomac.2023.124490] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 04/05/2023] [Accepted: 04/13/2023] [Indexed: 04/21/2023]
Abstract
Polysaccharides κ-carrageenan (κ-Car) have become a predominant source in developing bioactive materials. We aimed to develop biopolymer composite materials of κ-Car with coriander essential oil (CEO) (κ-Car-CEO) films for fibroblast-associated wound healing. Initially, we loaded the CEO in to κ-Car and CEO through homogenization and ultrasonication to fabricate composite film bioactive materials. After performing morphological and chemical characterizations, we validated the developed material functionalities in both in vitro and in vivo models. The chemical and morphological analysis with physical structure, swelling ratio, encapsulation efficiency, CEO release, and water barrier properties of films examined and showed the structural interaction of κ-Car and CEO-loaded into the polymer network. Furthermore, the bioactive applications of CEO release showed initial burst release followed by controlled release from the κ-Car composite film with fibroblast (L929) cell adhesive capabilities and mechanosensing. Our results proved that the CEO-loaded into the κ-Car film impacts cell adhesion, F-actin organization, and collagen synthesis, followed by in vitro mechanosensing activation, further promoting wound healing in vivo. Our innovative perspectives of active polysaccharide (κ-Car)-based CEO functional film materials could potentially accomplish regenerative medicine.
Collapse
Affiliation(s)
- Malairaj Sathuvan
- Department of Biology & Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, College of Science, Shantou University, Shantou, Guangdong 515063, PR China
| | - Ramar Thangam
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea; Institute for High Technology Materials and Devices, Korea University, Seoul 02841, Republic of Korea
| | - Kit-Leong Cheong
- Department of Biology & Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, College of Science, Shantou University, Shantou, Guangdong 515063, PR China
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Yang Liu
- Department of Biology & Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, College of Science, Shantou University, Shantou, Guangdong 515063, PR China.
| |
Collapse
|
58
|
Jin X, Jiang C, Zou Z, Huang H, Li X, Xu S, Tan R. Ferritinophagy in the etiopathogenic mechanism of related diseases. J Nutr Biochem 2023; 117:109339. [PMID: 37061010 DOI: 10.1016/j.jnutbio.2023.109339] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 03/18/2023] [Accepted: 03/23/2023] [Indexed: 04/17/2023]
Abstract
Iron is an essential trace element that is involved in a variety of physiological processes. Ferritinophagy is selective autophagy mediated by nuclear receptor coactivator 4 (NCOA4), which regulates iron homeostasis in the body. Upon iron depletion or starvation, ferritinophagy is activated, releasing large amounts of Fe2+ and increasing reactive oxygen species (ROS), leading to ferroptosis. This plays a significant role in the etiopathogenesis of many diseases, such as metabolic diseases, neurodegenerative diseases, infectious diseases, tumors, cardiomyopathy, and ischemia-reperfusion ischemia-reperfusion injury. Here, we first review the regulation and functions of ferritinophagy and then describe its involvement in different diseases, with hopes of providing new understanding and insights into iron metabolism and iron disorder-related diseases and the therapeutic opportunity for targeting ferritinophagy.
Collapse
Affiliation(s)
- Xuemei Jin
- Department of Preventive Medicine, School of Medicine, Yanbian University, Yanji, China; Department of Clinical Nutrition, Guangzhou Institute of Disease-Oriented Nutritional Research, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Chunjie Jiang
- Department of Clinical Nutrition, Guangzhou Institute of Disease-Oriented Nutritional Research, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Zhizhou Zou
- Department of Preventive Medicine, School of Medicine, Yanbian University, Yanji, China; Department of Clinical Nutrition, Guangzhou Institute of Disease-Oriented Nutritional Research, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - He Huang
- Department of Preventive Medicine, School of Medicine, Yanbian University, Yanji, China; Department of Clinical Nutrition, Guangzhou Institute of Disease-Oriented Nutritional Research, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Xiaojian Li
- Department of Burn, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Songji Xu
- Department of Preventive Medicine, School of Medicine, Yanbian University, Yanji, China
| | - Rongshao Tan
- Department of Clinical Nutrition, Guangzhou Institute of Disease-Oriented Nutritional Research, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China.
| |
Collapse
|
59
|
Wang J, Qiao Q, Sun Y, Yu W, Wang J, Zhu M, Yang K, Huang X, Bai Y. Osteogenic Differentiation Effect of Human Periodontal Ligament Stem-Cell Initial Cell Density on Autologous Cells and Human Bone Marrow Stromal Cells. Int J Mol Sci 2023; 24:ijms24087133. [PMID: 37108296 PMCID: PMC10138982 DOI: 10.3390/ijms24087133] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/02/2023] [Accepted: 04/05/2023] [Indexed: 04/29/2023] Open
Abstract
Stem cells have differentiation and regulation functions. Here, we discussed the impact of cell culture density on stem cell proliferation, osteoblastogenesis, and regulation. To discuss the effect of the initial culture density of human periodontal ligament stem cells (hPDLSCs) on the osteogenic differentiation of autologous cells, we found that the hPDLSC proliferation rate decreased with an increase in the initial plating density (0.5-8 × 104 cells/cm2) for the 48 h culture cycle. After hPDLSCs induced osteogenic differentiation for 14 days with different initial cell culture densities, the expression of osteoprotegerin (OPG) and runt-related transcription factor 2(RUNX2) and the OPG/ Receptor Activator of Nuclear Factor-κ B Ligand (RANKL) ratio were the highest in the hPDLSCs initially plated at a density of 2 × 104 cells/cm2, and the average cell calcium concentration was also the highest. To study hPDLSCs regulating the osteoblastic differentiation of other cells, we used 50 μg/mL of secreted exosomes derived from hPDLSCs cultured using different initial cell densities to induce human bone marrow stromal cell (hBMSC) osteogenesis. After 14 days, the results indicated that the gene expression of OPG, Osteocalcin(OCN,)RUNX2, and osterix and the OPG/RANKL ratio were the highest in the 2 × 104 cells/cm2 initial cell density group, and the average calcium concentration was also the highest. This provides a new idea for the clinical application of stem cell osteogenesis.
Collapse
Affiliation(s)
- Jing Wang
- Department of Orthodontics, School of Stomatology, Beijing Stomatological Hospital, Capital Medical University, Beijing 100050, China
| | - Qingchen Qiao
- Department of Orthodontics, School of Stomatology, Beijing Stomatological Hospital, Capital Medical University, Beijing 100050, China
| | - Yaxi Sun
- Department of Orthodontics, School of Stomatology, Beijing Stomatological Hospital, Capital Medical University, Beijing 100050, China
| | - Wenting Yu
- Department of Orthodontics, School of Stomatology, Beijing Stomatological Hospital, Capital Medical University, Beijing 100050, China
| | - Jiran Wang
- Department of Orthodontics, School of Stomatology, Beijing Stomatological Hospital, Capital Medical University, Beijing 100050, China
| | - Minjia Zhu
- Department of Orthodontics, School of Stomatology, Beijing Stomatological Hospital, Capital Medical University, Beijing 100050, China
| | - Kai Yang
- Department of Orthodontics, School of Stomatology, Beijing Stomatological Hospital, Capital Medical University, Beijing 100050, China
| | - Xiaofeng Huang
- Department of Stomatology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Yuxing Bai
- Department of Orthodontics, School of Stomatology, Beijing Stomatological Hospital, Capital Medical University, Beijing 100050, China
| |
Collapse
|
60
|
Matarrese P, Vona R, Ascione B, Cittadini C, Tocci A, Mileo AM. Tumor Microenvironmental Cytokines Drive NSCLC Cell Aggressiveness and Drug-Resistance via YAP-Mediated Autophagy. Cells 2023; 12:cells12071048. [PMID: 37048121 PMCID: PMC10093141 DOI: 10.3390/cells12071048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 03/27/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
Dynamic reciprocity between cellular components of the tumor microenvironment and tumor cells occurs primarily through the interaction of soluble signals, i.e., cytokines produced by stromal cells to support cancer initiation and progression by regulating cell survival, differentiation and immune cell functionality, as well as cell migration and death. In the present study, we focused on the analysis of the functional response of non-small cell lung cancer cell lines elicited by the treatment with some crucial stromal factors which, at least in part, mimic the stimulus exerted in vivo on tumor cells by microenvironmental components. Our molecular and functional results highlight the role played by the autophagic machinery in the cellular response in terms of the invasive capacity, stemness and drug resistance of two non-small lung cancer cell lines treated with stromal cytokines, also highlighting the emerging role of the YAP pathway in the mutual and dynamic crosstalk between tumor cells and tumor microenvironment elements. The results of this study provide new insights into the YAP-mediated autophagic mechanism elicited by microenvironmental cytokines on non-small cell lung cancer cell lines and may suggest new potential strategies for future cancer therapeutic interventions.
Collapse
Affiliation(s)
- Paola Matarrese
- Oncology Unit, Center for Gender-Specific Medicine, Italian National Institute of Health, Viale Regina Elena, 299-00161 Rome, Italy
- Correspondence: (P.M.); (A.M.M.)
| | - Rosa Vona
- Oncology Unit, Center for Gender-Specific Medicine, Italian National Institute of Health, Viale Regina Elena, 299-00161 Rome, Italy
| | - Barbara Ascione
- Oncology Unit, Center for Gender-Specific Medicine, Italian National Institute of Health, Viale Regina Elena, 299-00161 Rome, Italy
| | - Camilla Cittadini
- Oncology Unit, Center for Gender-Specific Medicine, Italian National Institute of Health, Viale Regina Elena, 299-00161 Rome, Italy
| | - Annalisa Tocci
- Tumor Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53-00144 Rome, Italy
| | - Anna Maria Mileo
- Tumor Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53-00144 Rome, Italy
- Correspondence: (P.M.); (A.M.M.)
| |
Collapse
|
61
|
Shi P, Cheng Z, Zhao K, Chen Y, Zhang A, Gan W, Zhang Y. Active targeting schemes for nano-drug delivery systems in osteosarcoma therapeutics. J Nanobiotechnology 2023; 21:103. [PMID: 36944946 PMCID: PMC10031984 DOI: 10.1186/s12951-023-01826-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 02/18/2023] [Indexed: 03/23/2023] Open
Abstract
Osteosarcoma, the most common malignant tumor of the bone, seriously influences people's lives and increases their economic burden. Conventional chemotherapy drugs achieve limited therapeutic effects owing to poor targeting and severe systemic toxicity. Nanocarrier-based drug delivery systems can significantly enhance the utilization efficiency of chemotherapeutic drugs through targeting ligand modifications and reduce the occurrence of systemic adverse effects. A variety of ligand-modified nano-drug delivery systems have been developed for different targeting schemes. Here we review the biological characteristics and the main challenges of current drug therapy of OS, and further elaborate on different targeting schemes and ligand selection for nano-drug delivery systems of osteosarcoma, which may provide new horizons for the development of advanced targeted drug delivery systems in the future.
Collapse
Affiliation(s)
- Pengzhi Shi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhangrong Cheng
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Kangcheng Zhao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yuhang Chen
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Anran Zhang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Weikang Gan
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yukun Zhang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
62
|
Hashemi M, Paskeh MDA, Orouei S, Abbasi P, Khorrami R, Dehghanpour A, Esmaeili N, Ghahremanzade A, Zandieh MA, Peymani M, Salimimoghadam S, Rashidi M, Taheriazam A, Entezari M, Hushmandi K. Towards dual function of autophagy in breast cancer: A potent regulator of tumor progression and therapy response. Biomed Pharmacother 2023; 161:114546. [PMID: 36958191 DOI: 10.1016/j.biopha.2023.114546] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/11/2023] [Accepted: 03/14/2023] [Indexed: 03/25/2023] Open
Abstract
As a devastating disease, breast cancer has been responsible for decrease in life expectancy of females and its morbidity and mortality are high. Breast cancer is the most common tumor in females and its treatment has been based on employment of surgical resection, chemotherapy and radiotherapy. The changes in biological behavior of breast tumor relies on genomic and epigenetic mutations and depletions as well as dysregulation of molecular mechanisms that autophagy is among them. Autophagy function can be oncogenic in increasing tumorigenesis, and when it has pro-death function, it causes reduction in viability of tumor cells. The carcinogenic function of autophagy in breast tumor is an impediment towards effective therapy of patients, as it can cause drug resistance and radio-resistance. The important hallmarks of breast tumor such as glucose metabolism, proliferation, apoptosis and metastasis can be regulated by autophagy. Oncogenic autophagy can inhibit apoptosis, while it promotes stemness of breast tumor. Moreover, autophagy demonstrates interaction with tumor microenvironment components such as macrophages and its level can be regulated by anti-tumor compounds in breast tumor therapy. The reasons of considering autophagy in breast cancer therapy is its pleiotropic function, dual role (pro-survival and pro-death) and crosstalk with important molecular mechanisms such as apoptosis. Moreover, current review provides a pre-clinical and clinical evaluation of autophagy in breast tumor.
Collapse
Affiliation(s)
- Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mahshid Deldar Abad Paskeh
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Sima Orouei
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Pegah Abbasi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Ramin Khorrami
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Amir Dehghanpour
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Negin Esmaeili
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Azin Ghahremanzade
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mohammad Arad Zandieh
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Maryam Peymani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari 4815733971, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari 4815733971, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| |
Collapse
|
63
|
Lee J, Roh JL. Epithelial-Mesenchymal Plasticity: Implications for Ferroptosis Vulnerability and Cancer Therapy. Crit Rev Oncol Hematol 2023; 185:103964. [PMID: 36931615 DOI: 10.1016/j.critrevonc.2023.103964] [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: 02/09/2023] [Revised: 03/12/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023] Open
Abstract
Cancers polarized to a mesenchymal or poorly differentiated state can often evade cell death induced by conventional therapies. The epithelial-mesenchymal transition is involved in lipid metabolism and increases polyunsaturated fatty acid levels in cancer cells, contributing to chemo- and radio-resistance. Altered metabolism in cancer enables invasion and metastasis but is prone to lipid peroxidation under oxidative stress. Cancers with mesenchymal rather than epithelial signatures are highly vulnerable to ferroptosis. Therapy-resistant persister cancer cells show a high mesenchymal cell state and dependence on the lipid peroxidase pathway, which can respond more sensitively to ferroptosis inducers. Cancer cells may survive under specific metabolic and oxidative stress conditions, and targeting this unique defense system can selectively kill only cancer cells. Therefore, this article summarizes the core regulatory mechanisms of ferroptosis in cancer, the relationship between ferroptosis and epithelial-mesenchymal plasticity, and the implications of epithelial-mesenchymal transition for ferroptosis-based cancer therapy.
Collapse
Affiliation(s)
- Jaewang Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea; Department of Biomedical Science, General Graduate School, CHA University, Seongnam, Republic of Korea
| | - Jong-Lyel Roh
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea; Department of Biomedical Science, General Graduate School, CHA University, Seongnam, Republic of Korea.
| |
Collapse
|
64
|
Yuan Z, Li Y, Zhang S, Wang X, Dou H, Yu X, Zhang Z, Yang S, Xiao M. Extracellular matrix remodeling in tumor progression and immune escape: from mechanisms to treatments. Mol Cancer 2023; 22:48. [PMID: 36906534 PMCID: PMC10007858 DOI: 10.1186/s12943-023-01744-8] [Citation(s) in RCA: 154] [Impact Index Per Article: 154.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/11/2023] [Indexed: 03/13/2023] Open
Abstract
The malignant tumor is a multi-etiological, systemic and complex disease characterized by uncontrolled cell proliferation and distant metastasis. Anticancer treatments including adjuvant therapies and targeted therapies are effective in eliminating cancer cells but in a limited number of patients. Increasing evidence suggests that the extracellular matrix (ECM) plays an important role in tumor development through changes in macromolecule components, degradation enzymes and stiffness. These variations are under the control of cellular components in tumor tissue via the aberrant activation of signaling pathways, the interaction of the ECM components to multiple surface receptors, and mechanical impact. Additionally, the ECM shaped by cancer regulates immune cells which results in an immune suppressive microenvironment and hinders the efficacy of immunotherapies. Thus, the ECM acts as a barrier to protect cancer from treatments and supports tumor progression. Nevertheless, the profound regulatory network of the ECM remodeling hampers the design of individualized antitumor treatment. Here, we elaborate on the composition of the malignant ECM, and discuss the specific mechanisms of the ECM remodeling. Precisely, we highlight the impact of the ECM remodeling on tumor development, including proliferation, anoikis, metastasis, angiogenesis, lymphangiogenesis, and immune escape. Finally, we emphasize ECM "normalization" as a potential strategy for anti-malignant treatment.
Collapse
Affiliation(s)
- Zhennan Yuan
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Yingpu Li
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Sifan Zhang
- Department of Neurobiology, Harbin Medical University, Harbin, 150081, China
| | - Xueying Wang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - He Dou
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Xi Yu
- Department of Gynecological Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Zhiren Zhang
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.,Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang Key Laboratory for Metabolic Disorder and Cancer Related Cardiovascular Diseases, Harbin, 150001, China
| | - Shanshan Yang
- Department of Gynecological Radiotherapy, Harbin Medical University Cancer Hospital, Harbin, 150000, China.
| | - Min Xiao
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China.
| |
Collapse
|
65
|
Johnson MS, Cook JG. Cell cycle exits and U-turns: Quiescence as multiple reversible forms of arrest. Fac Rev 2023; 12:5. [PMID: 36923701 PMCID: PMC10009890 DOI: 10.12703/r/12-5] [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] [Indexed: 03/10/2023] Open
Abstract
Cell proliferation control is essential during development and for maintaining adult tissues. Loss of that control promotes not only oncogenesis when cells proliferate inappropriately but also developmental abnormalities or degeneration when cells fail to proliferate when and where needed. To ensure that cells are produced at the right place and time, an intricate balance of pro-proliferative and anti-proliferative signals impacts the probability that cells undergo cell cycle exit to quiescence, or G0 phase. This brief review describes recent advances in our understanding of how and when quiescence is initiated and maintained in mammalian cells. We highlight the growing appreciation for quiescence as a collection of context-dependent distinct states.
Collapse
Affiliation(s)
- Martha Sharisha Johnson
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, NC, USA
| | - Jeanette Gowen Cook
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, NC, USA
| |
Collapse
|
66
|
Murakami K, Furuya H, Hokutan K, Goodison S, Pagano I, Chen R, Shen CH, Chan MWY, Ng CF, Kobayashi T, Ogawa O, Miyake M, Thornquist M, Shimizu Y, Hayashi K, Wang Z, Yu H, Rosser CJ. Association of SNPs in the PAI1 Gene with Disease Recurrence and Clinical Outcome in Bladder Cancer. Int J Mol Sci 2023; 24:4943. [PMID: 36902377 PMCID: PMC10003630 DOI: 10.3390/ijms24054943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
PURPOSE Bladder cancer (BCa) is one of the most common cancer types worldwide and is characterized by a high rate of recurrence. In previous studies, we and others have described the functional influence of plasminogen activator inhibitor-1 (PAI1) in bladder cancer development. While polymorphisms in PAI1 have been associated with increased risk and worsened prognosis in some cancers, the mutational status of PAI1 in human bladder tumors has not been well defined. METHODS In this study, we evaluated the mutational status of PAI1 in a series of independent cohorts, comprised of a total of 660 subjects. RESULTS Sequencing analyses identified two clinically relevant 3' untranslated region (UTR) single nucleotide polymorphisms (SNPs) in PAI1 (rs7242; rs1050813). Somatic SNP rs7242 was present in human BCa cohorts (overall incidence of 72%; 62% in Caucasians and 72% in Asians). In contrast, the overall incidence of germline SNP rs1050813 was 18% (39% in Caucasians and 6% in Asians). Furthermore, Caucasian patients with at least one of the described SNPs had worse recurrence-free survival and overall survival (p = 0.03 and p = 0.03, respectively). In vitro functional studies demonstrated that SNP rs7242 increased the anti-apoptotic effect of PAI1, and SNP rs1050813 was related to a loss of contact inhibition associated with cellular proliferation when compared to wild type. CONCLUSION Further investigation of the prevalence and potential downstream influence of these SNPs in bladder cancer is warranted.
Collapse
Affiliation(s)
- Kaoru Murakami
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Hideki Furuya
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Kanani Hokutan
- Clinical and Translational Research Program, University of Hawaii Cancer Center, Honolulu, HI 96813, USA
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu, HI 96822, USA
| | - Steve Goodison
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Ian Pagano
- Population Sciences in the Pacific Program, University of Hawaii Cancer Center, Honolulu, HI 96813, USA
| | - Runpu Chen
- Department of Microbiology and Immunology, The State University of New York at Buffalo, Buffalo, NY 14260, USA
| | - Cheng-Huang Shen
- Department of Urology, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 600, Taiwan
| | - Michael W. Y. Chan
- Department of Biomedical Sciences, National Chung Cheng University, Chia-Yi 621, Taiwan
| | - Chi Fai Ng
- SH Ho Urology Centre, Department of Surgery, The Chinese University of Hong Kong, Hong Kong
| | - Takashi Kobayashi
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Osamu Ogawa
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Makito Miyake
- Department of Urology, Nara Medical University, Nara 6348522, Japan
| | - Mark Thornquist
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Yoshiko Shimizu
- Clinical and Translational Research Program, University of Hawaii Cancer Center, Honolulu, HI 96813, USA
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu, HI 96822, USA
| | - Kazukuni Hayashi
- Clinical and Translational Research Program, University of Hawaii Cancer Center, Honolulu, HI 96813, USA
| | - Zhangwei Wang
- Population Sciences in the Pacific Program, University of Hawaii Cancer Center, Honolulu, HI 96813, USA
| | - Herbert Yu
- Population Sciences in the Pacific Program, University of Hawaii Cancer Center, Honolulu, HI 96813, USA
| | - Charles J. Rosser
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
67
|
Karrobi K, Tank A, Fuzail MA, Kalidoss M, Tilbury K, Zaman M, Ferruzzi J, Roblyer D. Fluorescence Lifetime Imaging Microscopy (FLIM) reveals spatial-metabolic changes in 3D breast cancer spheroids. Sci Rep 2023; 13:3624. [PMID: 36869092 PMCID: PMC9984376 DOI: 10.1038/s41598-023-30403-7] [Citation(s) in RCA: 7] [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: 11/20/2022] [Accepted: 02/22/2023] [Indexed: 03/05/2023] Open
Abstract
Cancer cells are mechanically sensitive to physical properties of the microenvironment, which can affect downstream signaling to promote malignancy, in part through the modulation of metabolic pathways. Fluorescence Lifetime Imaging Microscopy (FLIM) can be used to measure the fluorescence lifetime of endogenous fluorophores, such as the metabolic co-factors NAD(P)H and FAD, in live samples. We used multiphoton FLIM to investigate the changes in cellular metabolism of 3D breast spheroids derived from MCF-10A and MD-MB-231 cell lines embedded in collagen with varying densities (1 vs. 4 mg/ml) over time (Day 0 vs. Day 3). MCF-10A spheroids demonstrated spatial gradients, with the cells closest to the spheroid edge exhibiting FLIM changes consistent with a shift towards oxidative phosphorylation (OXPHOS) while the spheroid core had changes consistent with a shift towards glycolysis. The MDA-MB-231 spheroids had a large shift consistent with increased OXPHOS with a more pronounced change at the higher collagen concentration. The MDA-MB-231 spheroids invaded into the collagen gel over time and cells that traveled the farthest had the largest changes consistent with a shift towards OXPHOS. Overall, these results suggest that the cells in contact with the extracellular matrix (ECM) and those that migrated the farthest had changes consistent with a metabolic shift towards OXPHOS. More generally, these results demonstrate the ability of multiphoton FLIM to characterize how spheroids metabolism and spatial metabolic gradients are modified by physical properties of the 3D ECM.
Collapse
Affiliation(s)
- Kavon Karrobi
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
| | - Anup Tank
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
| | | | - Madhumathi Kalidoss
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
| | - Karissa Tilbury
- Department of Chemical and Biomedical Engineering, University of Maine, Orono, ME, 04469, USA
| | - Muhammad Zaman
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
| | - Jacopo Ferruzzi
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
- Department of Biomedical Engineering, The University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Darren Roblyer
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA.
| |
Collapse
|
68
|
Choi JH, Park SY, Lee WJ, Lee CJ, Kim JH, Jang TY, Jeon SE, Jun Y, Nam JS. SEC22B inhibition attenuates colorectal cancer aggressiveness and autophagic flux under unfavorable environment. Biochem Biophys Res Commun 2023; 665:10-18. [PMID: 37148741 DOI: 10.1016/j.bbrc.2023.03.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/13/2023] [Accepted: 03/14/2023] [Indexed: 03/17/2023]
Abstract
Autophagy has bidirectional functions in cancer by facilitating cell survival and death in a context-dependent manner. Soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs) are a large family of proteins essential for numerous biological processes, including autophagy; nevertheless, their potential function in cancer malignancy remains unclear. Here, we explored the gene expression patterns of SNAREs in tissues of patients with colorectal cancer (CRC) and discovered that SEC22B expression, a vesicle SNARE, was higher in tumor tissues than in normal tissues, with a more significant increase in metastatic tissues. Interestingly, SEC22B knockdown dramatically decreased CRC cell survival and growth, especially under stressful conditions, such as hypoxia and serum starvation, and decreased the number of stress-induced autophagic vacuoles. Moreover, SEC22B knockdown successfully attenuated liver metastasis in a CRC cell xenograft mouse model, with histological signs of decreased autophagic flux and proliferation within cancer cells. Together, this study posits that SEC22B plays a crucial role in enhancing the aggressiveness of CRC cells, suggesting that SEC22B might be an attractive therapeutic target for CRC.
Collapse
|
69
|
Alhasan B, Mikeladze M, Guzhova I, Margulis B. Autophagy, molecular chaperones, and unfolded protein response as promoters of tumor recurrence. Cancer Metastasis Rev 2023; 42:217-254. [PMID: 36723697 DOI: 10.1007/s10555-023-10085-3] [Citation(s) in RCA: 7] [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: 08/23/2022] [Accepted: 01/16/2023] [Indexed: 02/02/2023]
Abstract
Tumor recurrence is a paradoxical function of a machinery, whereby a small proportion of the cancer cell population enters a resistant, dormant state, persists long-term in this condition, and then transitions to proliferation. The dormant phenotype is typical of cancer stem cells, tumor-initiating cells, disseminated tumor cells, and drug-tolerant persisters, which all demonstrate similar or even equivalent properties. Cancer cell dormancy and its conversion to repopulation are regulated by several protein signaling systems that inhibit or induce cell proliferation and provide optimal interrelations between cancer cells and their special niche; these systems act in close connection with tumor microenvironment and immune response mechanisms. During dormancy and reawakening periods, cell proteostasis machineries, autophagy, molecular chaperones, and the unfolded protein response are recruited to protect refractory tumor cells from a wide variety of stressors and therapeutic insults. Proteostasis mechanisms functionally or even physically interfere with the main regulators of tumor relapse, and the significance of these interactions and implications in the tumor recurrence phases are discussed in this review.
Collapse
Affiliation(s)
- Bashar Alhasan
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064, St. Petersburg, Russia.
| | - Marina Mikeladze
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064, St. Petersburg, Russia
| | - Irina Guzhova
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064, St. Petersburg, Russia
| | - Boris Margulis
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064, St. Petersburg, Russia
| |
Collapse
|
70
|
Chien CY, Lin JC, Huang CY, Hsu CY, Yang KC, Chattopadhyay S, Nikoloutsos N, Hsieh PCH, Hu CMJ. In Situ Hydrogelation of Cellular Monolayers Enables Conformal Biomembrane Functionalization for Xeno-Free Feeder Substrate Engineering. Adv Healthc Mater 2023; 12:e2201708. [PMID: 36455286 DOI: 10.1002/adhm.202201708] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 11/14/2022] [Indexed: 12/03/2022]
Abstract
The intricate functionalities of cellular membranes have inspired strategies for deriving and anchoring cell-surface components onto solid substrates for biological studies, biosensor applications, and tissue engineering. However, introducing conformal and right-side-out cell membrane coverage onto planar substrates requires cumbersome protocols susceptible to significant device-to-device variability. Here, a facile approach for biomembrane functionalization of planar substrates is demonstrated by subjecting confluent cellular monolayer to intracellular hydrogel polymerization. The resulting cell-gel hybrid, herein termed GELL (gelated cell), exhibits extraordinary stability and retains the structural integrity, membrane fluidity, membrane protein mobility, and topology of living cells. In assessing the utility of GELL layers as a tissue engineering feeder substrate for stem cell maintenance, GELL feeder prepared from primary mouse embryonic fibroblasts not only preserves the stemness of murine stem cells but also exhibits advantages over live feeder cells owing to the GELL's inanimate, non-metabolizing nature. The preparation of a xeno-free feeder substrate devoid of non-human components is further shown with HeLa cells, and the resulting HeLa GELL feeder effectively sustains the growth and stemness of both murine and human induced pluripotent stem cells. The study highlights a novel bio-functionalization strategy that introduces new opportunities for tissue engineering and other biomedical applications.
Collapse
Affiliation(s)
- Chen-Ying Chien
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - Jung-Chen Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
- Institute of Oral Biology, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Ching-Ying Huang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - Chung-Yao Hsu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academic Sinica, Taipei, 11529, Taiwan
| | - Kai-Chieh Yang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academic Sinica, Taipei, 11529, Taiwan
| | - Saborni Chattopadhyay
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academic Sinica, Taipei, 11529, Taiwan
| | | | | | - Che-Ming Jack Hu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academic Sinica, Taipei, 11529, Taiwan
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan, 701, Taiwan
| |
Collapse
|
71
|
New Sources, Differentiation, and Therapeutic Uses of Mesenchymal Stem Cells 2.0. Int J Mol Sci 2023; 24:ijms24043938. [PMID: 36835347 PMCID: PMC9963112 DOI: 10.3390/ijms24043938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 02/08/2023] [Indexed: 02/18/2023] Open
Abstract
For the clinical application of mesenchymal stem cells (MSCs), the optimization of biological products (e [...].
Collapse
|
72
|
Liu S, Li Y, Yuan M, Song Q, Liu M. Correlation between the Warburg effect and progression of triple-negative breast cancer. Front Oncol 2023; 12:1060495. [PMID: 36776368 PMCID: PMC9913723 DOI: 10.3389/fonc.2022.1060495] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 12/06/2022] [Indexed: 01/28/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is ineligible for hormonal therapy and Her-2-targeted therapy due to the negative expression of the estrogen receptor, progesterone receptor, and human epidermal growth factor receptor-2. Although targeted therapy and immunotherapy have been shown to attenuate the aggressiveness of TNBC partially, few patients have benefited from them. The conventional treatment for TNBC remains chemotherapy. Chemoresistance, however, impedes therapeutic progress over time, and chemotherapy toxicity increases the burden of cancer on patients. Therefore, introducing more advantageous TNBC treatment options is a necessity. Metabolic reprogramming centered on glucose metabolism is considered a hallmark of tumors. It is described as tumor cells tend to convert glucose to lactate even under normoxic conditions, a phenomenon known as the Warburg effect. Similar to Darwinian evolution, its emergence is attributed to the selective pressures formed by the hypoxic microenvironment of pre-malignant lesions. Of note, the Warburg effect does not disappear with changes in the microenvironment after the formation of malignant tumor phenotypes. Instead, it forms a constitutive expression mediated by mutations or epigenetic modifications, providing a robust selective survival advantage for primary and metastatic lesions. Expanding evidence has demonstrated that the Warburg effect mediates multiple invasive behaviors in TNBC, including proliferation, metastasis, recurrence, immune escape, and multidrug resistance. Moreover, the Warburg effect-targeted therapy has been testified to be feasible in inhibiting TNBC progression. However, not all TNBCs are sensitive to glycolysis inhibitors because TNBC cells flexibly switch their metabolic patterns to cope with different survival pressures, namely metabolic plasticity. Between the Warburg effect-targeted medicines and the actual curative effect, metabolic plasticity creates a divide that must be continuously researched and bridged.
Collapse
Affiliation(s)
| | | | | | - Qing Song
- *Correspondence: Min Liu, ; Qing Song,
| | - Min Liu
- *Correspondence: Min Liu, ; Qing Song,
| |
Collapse
|
73
|
Carvalho J. A computational model of cell membrane bioelectric polarization and depolarization, connected with cell proliferation, in different tissue geometries. J Theor Biol 2023; 557:111338. [PMID: 36343668 DOI: 10.1016/j.jtbi.2022.111338] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 08/23/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022]
Abstract
A reliable theory of biological tissues growth and organization, a fundamental tool for a comprehensive interpretation of experimental observations and a guide to progress in life sciences, is definitively missing. This would support the advancement of knowledge for both normal and pathological expansion and regulation of tissues and organisms. In this work is presented a computational model of cell culture that describes its growth and organization using cell proliferation as its default state, constrained by contact inhibition, closely connected to the cell bioelectric state. The model results describe in a correct way the reported experimental results, involving contact inhibition due to the presence of other cells, and gap junctions for signaling, molecules exchange and extracellular environment sensing. Starting from depolarized cells (in this model considered tantamount to proliferative), the cell culture grows until it fills the available domain and, due to the contact inhibition constraint, it turns into quiescence (a consequence of cell polarization), except on the periphery. Using drugs or via protein expression manipulation, it is possible to change the final tissue state, to fully polarized or depolarized. Other experimental tests are proposed and the expected results simulated. This model can be extended to pathological events, such as carcinogenesis, with cells homeostasis perturbed by a cell depolarizing (carcinogenic) event and express its default proliferative state without adequate control. This simplified model of tissue organization, regulated by the cell's bioelectric state and their interaction with vicinity, is an alternative to the description of the experimental results by mechanical stress, and can be further tested and extended in dedicated experiments.
Collapse
Affiliation(s)
- Joao Carvalho
- CFisUC, Department of Physics, University of Coimbra, Portugal.
| |
Collapse
|
74
|
Maejima Y, Zablocki D, Nah J, Sadoshima J. The role of the Hippo pathway in autophagy in the heart. Cardiovasc Res 2023; 118:3320-3330. [PMID: 35150237 DOI: 10.1093/cvr/cvac014] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 02/07/2022] [Indexed: 01/25/2023] Open
Abstract
The Hippo pathway, an evolutionarily conserved signalling mechanism, controls organ size and tumourigenesis. Increasing lines of evidence suggest that autophagy, an important mechanism of lysosome-mediated cellular degradation, is regulated by the Hippo pathway, which thereby profoundly affects cell growth and death responses in various cell types. In the heart, Mst1, an upstream component of the Hippo pathway, not only induces apoptosis but also inhibits autophagy through phosphorylation of Beclin 1. YAP/TAZ, transcription factor co-factors and the terminal effectors of the Hippo pathway, affect autophagy through transcriptional activation of TFEB, a master regulator of autophagy and lysosomal biogenesis. The cellular abundance of YAP is negatively regulated by autophagy and suppression of autophagy induces accumulation of YAP, which, in turn, acts as a feedback mechanism to induce autophagosome formation. Thus, the Hippo pathway and autophagy regulate each other, thereby profoundly affecting cardiomyocyte survival and death. This review discusses the interaction between the Hippo pathway and autophagy and its functional significance during stress conditions in the heart and the cardiomyocytes therein.
Collapse
Affiliation(s)
- Yasuhiro Maejima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, 185 South Orange Ave., MSB G-609, Newark, NJ 07103, USA.,Department of Cardiovascular Medicine, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Daniela Zablocki
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, 185 South Orange Ave., MSB G-609, Newark, NJ 07103, USA
| | - Jihoon Nah
- School of Biological Sciences, Seoul National University, Seoul 08826, Korea
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, 185 South Orange Ave., MSB G-609, Newark, NJ 07103, USA
| |
Collapse
|
75
|
Zanoletti L, Valdata A, Nehlsen K, Faris P, Casali C, Cacciatore R, Sbarsi I, Carriero F, Arfini D, van Baarle L, De Simone V, Barbieri G, Raimondi E, May T, Moccia F, Bozzola M, Matteoli G, Comincini S, Manai F. Cytological, molecular, cytogenetic, and physiological characterization of a novel immortalized human enteric glial cell line. Front Cell Neurosci 2023; 17:1170309. [PMID: 37153631 PMCID: PMC10158601 DOI: 10.3389/fncel.2023.1170309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 03/22/2023] [Indexed: 05/10/2023] Open
Abstract
Enteric glial cells (EGCs), the major components of the enteric nervous system (ENS), are implicated in the maintenance of gut homeostasis, thereby leading to severe pathological conditions when impaired. However, due to technical difficulties associated with EGCs isolation and cell culture maintenance that results in a lack of valuable in vitro models, their roles in physiological and pathological contexts have been poorly investigated so far. To this aim, we developed for the first time, a human immortalized EGC line (referred as ClK clone) through a validated lentiviral transgene protocol. As a result, ClK phenotypic glial features were confirmed by morphological and molecular evaluations, also providing the consensus karyotype and finely mapping the chromosomal rearrangements as well as HLA-related genotypes. Lastly, we investigated the ATP- and acetylcholine, serotonin and glutamate neurotransmitters mediated intracellular Ca2+ signaling activation and the response of EGCs markers (GFAP, SOX10, S100β, PLP1, and CCL2) upon inflammatory stimuli, further confirming the glial nature of the analyzed cells. Overall, this contribution provided a novel potential in vitro tool to finely characterize the EGCs behavior under physiological and pathological conditions in humans.
Collapse
Affiliation(s)
- Lisa Zanoletti
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Pavia, Italy
- Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven, Belgium
| | - Aurora Valdata
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Pavia, Italy
| | | | - Pawan Faris
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Pavia, Italy
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Iraq
| | - Claudio Casali
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Pavia, Italy
| | - Rosalia Cacciatore
- Immunohematology and Transfusion Service, I.R.C.C.S. Policlinico San Matteo, Pavia, Italy
| | - Ilaria Sbarsi
- Immunohematology and Transfusion Service, I.R.C.C.S. Policlinico San Matteo, Pavia, Italy
| | - Francesca Carriero
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Pavia, Italy
| | - Davide Arfini
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Pavia, Italy
| | - Lies van Baarle
- Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven, Belgium
| | - Veronica De Simone
- Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven, Belgium
| | - Giulia Barbieri
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Pavia, Italy
| | - Elena Raimondi
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Pavia, Italy
| | | | - Francesco Moccia
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Pavia, Italy
| | | | - Gianluca Matteoli
- Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven, Belgium
| | - Sergio Comincini
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Pavia, Italy
| | - Federico Manai
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Pavia, Italy
- *Correspondence: Federico Manai,
| |
Collapse
|
76
|
Vaughan-Jackson A, Stodolak S, Ebrahimi KH, Johnson E, Reardon PK, Dupont M, Zhang S, McCullagh JSO, James WS. Density dependent regulation of inflammatory responses in macrophages. Front Immunol 2022; 13:895488. [PMID: 36591218 PMCID: PMC9800520 DOI: 10.3389/fimmu.2022.895488] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022] Open
Abstract
Macrophage distribution density is tightly regulated within the body, yet the importance of macrophage crowding during in vitro culture is largely unstudied. Using a human induced pluripotent stem cell (iPSC)-derived macrophage model of tissue resident macrophages, we characterize how increasing macrophage culture density changes their morphology and phenotype before and after inflammatory stimulation. In particular, density drives changes in macrophage inflammatory cytokine and chemokine secretion in both resting and activated states. This density regulated inflammatory state is also evident in blood monocyte derived-macrophages, the human monocytic THP-1 immortalized cell line, and iPSC-derived microglia. Density-dependent changes appear to be driven by a transferable soluble factor, yet the precise mechanism remains unknown. Our findings highlight cell plating density as an important but frequently overlooked consideration of in vitro macrophage research relevant to a variety of fields ranging from basic macrophage cell biology to disease studies.
Collapse
Affiliation(s)
- Alun Vaughan-Jackson
- James & Lillian Martin Centre, Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Szymon Stodolak
- James & Lillian Martin Centre, Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | | | - Errin Johnson
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Paul K. Reardon
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
| | - Maeva Dupont
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Shengpan Zhang
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | | | - William S. James
- James & Lillian Martin Centre, Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
77
|
Xu L, Xiao T, Xu L, Yao W. Identification of therapeutic targets and prognostic biomarkers in cholangiocarcinoma via WGCNA. Front Oncol 2022; 12:977992. [PMID: 36591499 PMCID: PMC9795187 DOI: 10.3389/fonc.2022.977992] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 11/17/2022] [Indexed: 12/15/2022] Open
Abstract
Background Cholangiocarcinoma (CCA) is a highly aggressive malignant tumor for which limited treatment methods and prognostic signatures are available. This study aims to identify potential therapeutic targets and prognostic biomarkers for CCA. Methods Based on differentially expressed genes (DEGs) identified from The Cancer Genome Atlas (TCGA) data, our study identified key gene modules correlated with CCA patient survival by weighted gene coexpression network analysis (WGCNA). Cox regression analysis identified survival-related genes in the key gene modules. The biological properties of the survival-related genes were evaluated by CCK-8 and transwell assays. Then, these genes were used to construct a prognostic signature that was internally and externally validated. Additionally, by combining clinical characteristics with the gene-based prognostic signature, a nomogram for survival prediction was built. Results WGCNA divided the 1531 DEGs into four gene modules, and the yellow gene module was significantly associated with overall survival (OS) and histologic neoplasm grade. Our study identified the lncRNA AGAP2-AS1 and a novel gene, GOLGA7B, that are closely related to survival. GOLGA7B downregulation promoted the invasion, migration and proliferation of CCA cells, but AGAP2-AS1 had the opposite effect. AGAP2-AS1 and GOLGA7B were integrated into a gene-based prognostic signature, and both internal and external validation studies confirmed that this two-gene prognostic signature and nomogram could accurately predict CCA patient prognosis. Conclusion AGAP2-AS1 and GOLGA7B are potential therapeutic targets and prognostic biomarkers for CCA.
Collapse
Affiliation(s)
- Lei Xu
- Department of Pediatrics Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ting Xiao
- Department of Ultrasonography Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ling Xu
- Department of Nursing Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wei Yao
- Department of Oncology Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China,*Correspondence: Wei Yao,
| |
Collapse
|
78
|
Fu R, Jiang X, Li G, Zhu Y, Zhang H. Junctional complexes in epithelial cells: sentinels for extracellular insults and intracellular homeostasis. FEBS J 2022; 289:7314-7333. [PMID: 34453866 DOI: 10.1111/febs.16174] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 08/13/2021] [Accepted: 08/27/2021] [Indexed: 01/13/2023]
Abstract
The cell-cell and cell-ECM junctions within the epithelial tissues are crucial anchoring structures that provide architectural stability, mechanical resistance, and permeability control. Their indispensable role as signaling hubs orchestrating cell shape-related changes such as proliferation, differentiation, migration, and apoptosis has also been well recognized. However, growing amount of evidence now suggests that the multitasking nature of epithelial junctions extends well beyond anchorage-dependent or cell shape change-related biological processes. In this review, we discuss the emerging roles of junctional complexes in regulating innate immune defense, stress resistance, and intracellular proteostasis of the epithelial cells, with emphasis on the upstream regulation of epithelial junctions on various aspects of the epithelial barrier.
Collapse
Affiliation(s)
- Rong Fu
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, China
| | - Xiaowan Jiang
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, China
| | - Gang Li
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, China
| | - Yi Zhu
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, China
| | - Huimin Zhang
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, China
| |
Collapse
|
79
|
Lee M, Du H, Winer DA, Clemente-Casares X, Tsai S. Mechanosensing in macrophages and dendritic cells in steady-state and disease. Front Cell Dev Biol 2022; 10:1044729. [PMID: 36467420 PMCID: PMC9712790 DOI: 10.3389/fcell.2022.1044729] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 11/01/2022] [Indexed: 11/18/2022] Open
Abstract
Macrophages and dendritic cells are myeloid cells that play critical roles in immune responses. Macrophages help to maintain homeostasis through tissue regeneration and the clearance of dead cells, but also mediate inflammatory processes against invading pathogens. As the most potent antigen-presenting cells, dendritic cells are important in connecting innate to adaptive immune responses via activation of T cells, and inducing tolerance under physiological conditions. While it is known that macrophages and dendritic cells respond to biochemical cues in the microenvironment, the role of extracellular mechanical stimuli is becoming increasingly apparent. Immune cell mechanotransduction is an emerging field, where accumulating evidence suggests a role for extracellular physical cues coming from tissue stiffness in promoting immune cell recruitment, activation, metabolism and inflammatory function. Additionally, many diseases such as pulmonary fibrosis, cardiovascular disease, cancer, and cirrhosis are associated with changes to the tissue biophysical environment. This review will discuss current knowledge about the effects of biophysical cues including matrix stiffness, topography, and mechanical forces on macrophage and dendritic cell behavior under steady-state and pathophysiological conditions. In addition, we will also provide insight on molecular mediators and signaling pathways important in macrophage and dendritic cell mechanotransduction.
Collapse
Affiliation(s)
- Megan Lee
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Huixun Du
- Buck Institute for Research on Aging, Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, United States
| | - Daniel A. Winer
- Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Pathology, University Health Network, Toronto, ON, Canada
- Buck Institute for Research on Aging, Novato, CA, United States
| | - Xavier Clemente-Casares
- Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, AB, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| | - Sue Tsai
- Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, AB, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
80
|
YAP promotes cell-autonomous immune responses to tackle intracellular Staphylococcus aureus in vitro. Nat Commun 2022; 13:6995. [PMID: 36384856 PMCID: PMC9669043 DOI: 10.1038/s41467-022-34432-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 10/24/2022] [Indexed: 11/17/2022] Open
Abstract
Transcriptional cofactors YAP/TAZ have recently been found to support autophagy and inflammation, which are part of cell-autonomous immunity and are critical in antibacterial defense. Here, we studied the role of YAP against Staphylococcus aureus using CRISPR/Cas9-mutated HEK293 cells and a primary cell-based organoid model. We found that S. aureus infection increases YAP transcriptional activity, which is required to reduce intracellular S. aureus replication. A 770-gene targeted transcriptomic analysis revealed that YAP upregulates genes involved in autophagy/lysosome and inflammation pathways in both infected and uninfected conditions. The YAP-TEAD transcriptional activity promotes autophagic flux and lysosomal acidification, which are then important for defense against intracellular S. aureus. Furthermore, the staphylococcal toxin C3 exoenzyme EDIN-B was found effective in preventing YAP-mediated cell-autonomous immune response. This study provides key insights on the anti-S. aureus activity of YAP, which could be conserved for defense against other intracellular bacteria.
Collapse
|
81
|
Chen G, Chen S, Di X, He S, Liu Y, Qu R, Luo Y, Liu Y, Yang L. Survivin knockdown alleviates pathological hydrostatic pressure-induced bladder smooth muscle cell dysfunction and BOO-induced bladder remodeling via autophagy. Front Cell Dev Biol 2022; 10:999547. [PMID: 36393846 PMCID: PMC9649584 DOI: 10.3389/fcell.2022.999547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 10/10/2022] [Indexed: 09/05/2023] Open
Abstract
Aim: Bladder outlet obstruction (BOO) leads to bladder wall remodeling accompanying the progression from inflammation to fibrosis where pathological hydrostatic pressure (HP)-induced alteration of bladder smooth muscle cells (BSMCs) hypertrophic and excessive extracellular matrix (ECM) deposition play a pivotal role. Recently, we have predicted survivin (BIRC5) as a potential hub gene that might be critical during bladder fibrosis by bioinformatics analyses from rat BOO bladder, but its function during BOO progression remains unknown. Here, we investigated the role of survivin protein on bladder dysfunction of BOO both in vitro and in vivo. Methods: Sprague-Dawley female rats were divided into three groups: control group, BOO group, and BOO followed by the treatment with YM155 group. Bladder morphology and function were evaluated by Masson staining and urodynamic testing. To elucidate the underlying mechanism, hBSMCs were subjected to pathological HP of 200 cm H2O and co-cultured with the presence or absence of survivin siRNA and/or autophagy inhibitor 3-MA. Autophagy was evaluated by the detection of Beclin1 and LC3B-II expression, proliferation was conducted by the EdU analysis and PCNA expression, and fibrosis was assessed by the examination of Col 1 and Fn expression. Results: BOO led to a gradual alteration of hypertrophy and fibrosis of the bladder, and subsequently induced bladder dysfunction accompanied by increased survivin expression, while these histological and function changes were attenuated by the treatment with YM155. HP significantly increased survivin expression, upregulated Col1 and Fn expression, enhanced proliferation, and downregulated autophagy markers, but these changes were partially abolished by survivin siRNA treatment, which was consistent with the results of the BOO rat experiment. In addition, the anti-fibrotic and anti-proliferative effects of the survivin siRNA treatment on hBSMCs were diminished after the inhibition of autophagy by the treatment with 3-MA. Conclusion: In summary, the upregulation of survivin increased cell proliferation and fibrotic protein expression of hBSMC and drove the onset of bladder remodeling through autophagy during BOO. Targeting survivin in pathological hBSMCs could be a promising way to anti-fibrotic therapeutic approach in bladder remodeling secondary to BOO.
Collapse
Affiliation(s)
- Guo Chen
- Department of Urology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
- Laboratory of Reconstructive Urology, West China Hospital, Institute of Urology, Sichuan University, Chengdu, China
| | - Shuang Chen
- Laboratory of Reconstructive Urology, West China Hospital, Institute of Urology, Sichuan University, Chengdu, China
| | - Xingpeng Di
- Laboratory of Reconstructive Urology, West China Hospital, Institute of Urology, Sichuan University, Chengdu, China
| | - Shengyin He
- Department of Urology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yugao Liu
- Department of Urology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Rui Qu
- Department of Urology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yi Luo
- Department of Urology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yuebai Liu
- Department of Education and Training, Sichuan Cancer Hospital, Chengdu, China
| | - Luo Yang
- Department of Urology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
82
|
Ferrer-Torres D, Wu JH, Zhang CJ, Hammer MA, Dame MK, Wu A, Holloway EM, Karpoff K, McCarthy CL, Bohm MS, Cuttitta AJ, Tigani DJ, Huang S, Tsai YH, Miller AJ, Walker T, Bayer DE, Hogan SP, Turgeon DK, Lin J, Higgins PDR, Sexton J, Spence JR. Mapping the adult human esophagus in vivo and in vitro. Development 2022; 149:dev200614. [PMID: 36278875 PMCID: PMC9720751 DOI: 10.1242/dev.200614] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 07/20/2022] [Indexed: 10/22/2023]
Abstract
Many esophageal diseases can arise during development or throughout life. Therefore, well-characterized in vitro models and detailed methods are essential for studying human esophageal development, homeostasis and disease. Here, we (1) create an atlas of the cell types observed in the normal adult human esophagus; (2) establish an ancestrally diverse biobank of in vitro esophagus tissue to interrogate homeostasis and injury; and (3) benchmark in vitro models using the adult human esophagus atlas. We created a single-cell RNA sequencing reference atlas using fresh adult esophagus biopsies and a continuously expanding biobank of patient-derived in vitro cultures (n=55 lines). We identify and validate several transcriptionally distinct cell classes in the native human adult esophagus, with four populations belonging to the epithelial layer, including basal, epibasal, early differentiating and terminally differentiated luminal cells. Benchmarking in vitro esophagus cultures to the in vivo reference using single-cell RNA sequencing shows that the basal stem cells are robustly maintained in vitro, and the diversity of epithelial cell types in culture is dependent on cell density. We also demonstrate that cultures can be grown in 2D or as 3D organoids, and these methods can be employed for modeling the complete epithelial layers, thereby enabling in vitro modeling of the human adult esophagus.
Collapse
Affiliation(s)
- Daysha Ferrer-Torres
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Center for Cell Plasticity and Organ Design, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Joshua H. Wu
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Charles J. Zhang
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Max A. Hammer
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Michael K. Dame
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Angeline Wu
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Emily M. Holloway
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Kateryna Karpoff
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Caroline L. McCarthy
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Margaret S. Bohm
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Ashley J. Cuttitta
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Dominic J. Tigani
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Sha Huang
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Yu-Hwai Tsai
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Alyssa J. Miller
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Taylor Walker
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - David E. Bayer
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Simon P. Hogan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Danielle Kim Turgeon
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Jules Lin
- Department of Thoracic Surgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Peter D. R. Higgins
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Jonathan Sexton
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
- U-M Center for Drug Repurposing, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jason R. Spence
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Center for Cell Plasticity and Organ Design, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
83
|
Wang J, Zhu Q, Li R, Zhang J, Ye X, Li X. YAP1 protects against septic liver injury via ferroptosis resistance. Cell Biosci 2022; 12:163. [PMID: 36182901 PMCID: PMC9526934 DOI: 10.1186/s13578-022-00902-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 09/19/2022] [Indexed: 11/06/2022] Open
Abstract
Background The liver plays crucial roles in sepsis and is one of the major targets for sepsis-related injuries. Ferroptosis, a newly emerged form of lytic cell death, has been implicated in sepsis related organ failure. Yes-associated protein1 (YAP1), a key regulator of the Hippo signaling pathway, may be involved in ferroptosis development. This study aimed to elucidate the role of YAP1 in septic liver injury through regulating ferroptosis, especially ferritinophagy-mediated ferroptosis. Results Cecal ligation and puncture (CLP) models were constructed in control (Yap1flfl) and liver-conditional knockout mice (Yap1fl/fl Alb-Cre) to induce septic liver injury, while LO2 cells with or without YAP1 overexpression/deletion were stimulated by lipopolysaccharide (LPS) in vitro. Our study showed YAP1 knockdown aggravated CLP-induced liver injury and inflammation, as well as accelerated hepatocyte ferroptosis, revealed by down-regulated expression of GPX4, FTH1 and SLC7A11, along with up-regulated expression of SFXN1 and NCOA4. Consistently, YAP1 deficiency aggravated LO2 cells ferroptosis, but YAP1 overexpression alleviated LPS-induced LO2 ferritinophagy, as evidenced by reduced mitochondrial ROS and Fe2+, along with down-regulated expression of SFXN1 and NCOA4. Further co-IP assay verified that YAP1 disrupted the interaction between NCOA4 and FTH1, thus prevent the degradation of ferritin to Fe2+, further reduced the ROS production and suppressed ferroptosis. Conclusion YAP1 inhibits ferritinophagy-mediated ferroptosis in hepatocytes, and YAP1 deficiency aggravates sepsis-induced liver injury. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-022-00902-7.
Collapse
Affiliation(s)
- Jin Wang
- grid.413247.70000 0004 1808 0969Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei China
| | - Qian Zhu
- grid.413247.70000 0004 1808 0969Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei China
| | - Rui Li
- grid.413247.70000 0004 1808 0969Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei China
| | - Jing Zhang
- grid.413247.70000 0004 1808 0969Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei China
| | - Xujun Ye
- grid.413247.70000 0004 1808 0969Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei China
| | - Xinyi Li
- grid.413247.70000 0004 1808 0969Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei China
| |
Collapse
|
84
|
Tortorella I, Argentati C, Emiliani C, Morena F, Martino S. Biochemical Pathways of Cellular Mechanosensing/Mechanotransduction and Their Role in Neurodegenerative Diseases Pathogenesis. Cells 2022; 11:3093. [PMID: 36231055 PMCID: PMC9563116 DOI: 10.3390/cells11193093] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 12/11/2022] Open
Abstract
In this review, we shed light on recent advances regarding the characterization of biochemical pathways of cellular mechanosensing and mechanotransduction with particular attention to their role in neurodegenerative disease pathogenesis. While the mechanistic components of these pathways are mostly uncovered today, the crosstalk between mechanical forces and soluble intracellular signaling is still not fully elucidated. Here, we recapitulate the general concepts of mechanobiology and the mechanisms that govern the mechanosensing and mechanotransduction processes, and we examine the crosstalk between mechanical stimuli and intracellular biochemical response, highlighting their effect on cellular organelles' homeostasis and dysfunction. In particular, we discuss the current knowledge about the translation of mechanosignaling into biochemical signaling, focusing on those diseases that encompass metabolic accumulation of mutant proteins and have as primary characteristics the formation of pathological intracellular aggregates, such as Alzheimer's Disease, Huntington's Disease, Amyotrophic Lateral Sclerosis and Parkinson's Disease. Overall, recent findings elucidate how mechanosensing and mechanotransduction pathways may be crucial to understand the pathogenic mechanisms underlying neurodegenerative diseases and emphasize the importance of these pathways for identifying potential therapeutic targets.
Collapse
Affiliation(s)
- Ilaria Tortorella
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy
| | - Chiara Argentati
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy
| | - Carla Emiliani
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy
- Centro di Eccellenza CEMIN (Materiali Innovativi Nanostrutturali per Applicazioni Chimica Fisiche e Biomediche), University of Perugia, 06123 Perugia, Italy
| | - Francesco Morena
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy
| | - Sabata Martino
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy
- Centro di Eccellenza CEMIN (Materiali Innovativi Nanostrutturali per Applicazioni Chimica Fisiche e Biomediche), University of Perugia, 06123 Perugia, Italy
| |
Collapse
|
85
|
Zhou Y, Fang C, Xu H, Yuan L, Liu Y, Wang X, Zhang A, Shao A, Zhou D. Ferroptosis in glioma treatment: Current situation, prospects and drug applications. Front Oncol 2022; 12:989896. [PMID: 36249003 PMCID: PMC9557197 DOI: 10.3389/fonc.2022.989896] [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: 07/09/2022] [Accepted: 09/14/2022] [Indexed: 11/13/2022] Open
Abstract
Ferroptosis is a regulatory form of iron-dependent cell death caused by the accumulation of lipid-based reactive oxygen species (ROS) and differs from apoptosis, pyroptosis, and necrosis. Especially in neoplastic diseases, the susceptibility of tumor cells to ferroptosis affects prognosis and is associated with complex effects. Gliomas are the most common primary intracranial tumors, accounting for disease in 81% of patients with malignant brain tumors. An increasing number of studies have revealed the particular characteristics of iron metabolism in glioma cells. Therefore, agents that target a wide range of molecules involved in ferroptosis may regulate this process and enhance glioma treatment. Here, we review the underlying mechanisms of ferroptosis and summarize the potential therapeutic options for targeting ferroptosis in glioma.
Collapse
Affiliation(s)
- Yuhang Zhou
- Health Management Center, Tongde Hospital of Zhejiang Province, Hangzhou, China
- The First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Chaoyou Fang
- Department of Neurosurgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Houshi Xu
- Department of Neurosurgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ling Yuan
- Department of Neurosurgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yibo Liu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaoyu Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Anke Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: Anke Zhang, ; Anwen Shao, ; Danyang Zhou,
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: Anke Zhang, ; Anwen Shao, ; Danyang Zhou,
| | - Danyang Zhou
- Health Management Center, Tongde Hospital of Zhejiang Province, Hangzhou, China
- *Correspondence: Anke Zhang, ; Anwen Shao, ; Danyang Zhou,
| |
Collapse
|
86
|
A calpain-6/YAP axis in sarcoma stem cells that drives the outgrowth of tumors and metastases. Cell Death Dis 2022; 13:819. [PMID: 36153320 PMCID: PMC9509353 DOI: 10.1038/s41419-022-05244-3] [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: 01/28/2022] [Revised: 09/06/2022] [Accepted: 09/07/2022] [Indexed: 01/23/2023]
Abstract
Sarcomas include cancer stem cells, but how these cells contribute to local and metastatic relapse is largely unknown. We previously showed the pro-tumor functions of calpain-6 in sarcoma stem cells. Here, we use an osteosarcoma cell model, osteosarcoma tissues and transcriptomic data from human tumors to study gene patterns associated with calpain-6 expression or suppression. Calpain-6 modulates the expression of Hippo pathway genes and stabilizes the hippo effector YAP. It also modulates the vesicular trafficking of β-catenin degradation complexes. Calpain-6 expression is associated with genes of the G2M phase of the cell cycle, supports G2M-related YAP activities and up-regulated genes controlling mitosis in sarcoma stem cells and tissues. In mouse models of bone sarcoma, most tumor cells expressed calpain-6 during the early steps of tumor out-growth. YAP inhibition prevented the neoformation of primary tumors and metastases but had no effect on already developed tumors. It could even accelerate lung metastasis associated with large bone tumors by affecting tumor-associated inflammation in the host tissues. Our results highlight a specific mechanism involving YAP transcriptional activity in cancer stem cells that is crucial during the early steps of tumor and metastasis outgrowth and that could be targeted to prevent sarcoma relapse.
Collapse
|
87
|
The consequence of imine bond origination: Fabrication of rapid self-healing chitosan hydrogel as a drug delivery candidate for water-soluble drug. Eur Polym J 2022. [DOI: 10.1016/j.eurpolymj.2022.111605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
88
|
Liu G, Li J, Wu C. Reciprocal regulation of actin filaments and cellular metabolism. Eur J Cell Biol 2022; 101:151281. [PMID: 36343493 DOI: 10.1016/j.ejcb.2022.151281] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 09/23/2022] [Accepted: 10/25/2022] [Indexed: 12/14/2022] Open
Abstract
For cells to adhere, migrate and proliferate, remodeling of the actin cytoskeleton is required. This process consumes a large amount of ATP while having an intimate connection with cellular metabolism. Signaling pathways that regulate energy homeostasis can also affect actin dynamics, whereas a variety of actin binding proteins directly or indirectly interact with the anabolic and catabolic regulators in cells. Here, we discuss the inter-regulation between actin filaments and cellular metabolism, reviewing recent discoveries on key metabolic enzymes that respond to actin remodeling as well as historical findings on metabolic stress-induced cytoskeletal reorganization. We also address emerging techniques that would benefit the study of cytoskeletal dynamics and cellular metabolism in high spatial-temporal resolution.
Collapse
Affiliation(s)
- Geyao Liu
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Jiayi Li
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Congying Wu
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; International Cancer Institute, Peking University, Beijing 100191, China.
| |
Collapse
|
89
|
Zheng YY, Shi YF, Zhu HZ, Ding SJ, Zhou GH. Quality evaluation of cultured meat with plant protein scaffold. Food Res Int 2022; 161:111818. [DOI: 10.1016/j.foodres.2022.111818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/01/2022] [Accepted: 08/18/2022] [Indexed: 11/16/2022]
|
90
|
Zhang J, Zheng Y, Wang Y, Wang J, Sang A, Song X, Li X. YAP1 alleviates sepsis-induced acute lung injury via inhibiting ferritinophagy-mediated ferroptosis. Front Immunol 2022; 13:884362. [PMID: 35979359 PMCID: PMC9376389 DOI: 10.3389/fimmu.2022.884362] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 07/04/2022] [Indexed: 11/30/2022] Open
Abstract
Ferroptosis is a phospholipid peroxidation-mediated and iron-dependent cell death form, involved in sepsis-induced organ injury and other lung diseases. Yes-associated protein 1 (YAP1), a key regulator of the Hippo signaling pathway, could target multiple ferroptosis regulators. Herein, this study aimed to explore the involvement of ferroptosis in the etiopathogenesis of sepsis-induced acute lung injury (ALI) and demonstrate that YAP1 could disrupt ferritinophagy and moderate sepsis-induced ALI. Cecal ligation and puncture (CLP) models were constructed in wild-type (WT) and pulmonary epithelium-conditional knockout (YAP1f/f) mice to induce ALI, while MLE-12 cells with or without YAP1 overexpression were stimulated by lipopolysaccharide (LPS) in vitro. In-vivo modes showed that YAP1 knockout aggravated CLP-induced ALI and also accelerated pulmonary ferroptosis, as presented by the downregulated expression of GPX4, FTH1, and SLC7A11, along with the upregulated expression of SFXN1 and NCOA4. Transcriptome research identified these key genes and ferroptosis pathways involved in sepsis-induced ALI. In-vitro modes consistently verified that YAP1 deficiency boosted the ferrous iron accumulation and mitochondrial dysfunction in response to LPS. Furthermore, the co-IP assay revealed that YAP1 overexpression could prevent the degradation of ferritin to a mass of Fe2+ (ferritinophagy) via disrupting the NCOA4–FTH1 interaction, which blocked the transport of cytoplasmic Fe2+ into the mitochondria via the mitochondrial membrane protein (SFXN1), further reducing the generation of mitochondrial ROS. Therefore, these findings revealed that YAP1 could inhibit ferroptosis in a ferritinophagy-mediated manner, thus alleviating sepsis-induced ALI, which may provide a new approach to the therapeutic orientation for sepsis-induced ALI.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yongping Zheng
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yun Wang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jin Wang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Aming Sang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xuemin Song
- Research Centre of Anesthesiology and Critical Care Medicine, Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
- *Correspondence: Xuemin Song, ; Xinyi Li,
| | - Xinyi Li
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
- *Correspondence: Xuemin Song, ; Xinyi Li,
| |
Collapse
|
91
|
Barbeau S, Joushomme A, Chappe Y, Cardouat G, Baudrimont I, Freund-Michel V, Guibert C, Marthan R, Berger P, Vacher P, Percherancier Y, Quignard JF, Ducret T. Cell Confluence Modulates TRPV4 Channel Activity in Response to Hypoxia. Biomolecules 2022; 12:954. [PMID: 35883510 PMCID: PMC9313184 DOI: 10.3390/biom12070954] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/10/2022] [Accepted: 06/14/2022] [Indexed: 12/13/2022] Open
Abstract
Transient receptor potential vanilloid 4 (TRPV4) is a polymodal Ca2+-permeable channel involved in various hypoxia-sensitive pathophysiological phenomena. Different tools are available to study channel activity, requiring cells to be cultured at specific optimal densities. In the present study, we examined if cell density may influence the effect of hypoxia on TRPV4 activity. Transiently TRPV4-transfected HEK293T cells were seeded at low or high densities corresponding to non-confluent or confluent cells, respectively, on the day of experiments, and cultured under in vitro normoxia or hypoxia. TRPV4-mediated cytosolic Ca2+ responses, single-channel currents, and Ca2+ influx through the channel were measured using Ca2+ imaging/microspectrofluorimetric assay, patch-clamp, and Bioluminescence Resonance Energy Transfer (BRET), respectively. TRPV4 plasma membrane translocation was studied using confocal microscopy, biotinylation of cell surface proteins, and BRET. Our results show that hypoxia exposure has a differential effect on TRPV4 activation depending on cell confluence. At low confluence levels, TRPV4 response is increased in hypoxia, whereas at high confluence levels, TRPV4 response is strongly inhibited, due to channel internalization. Thus, cell density appears to be a crucial parameter for TRPV4 channel activity.
Collapse
Affiliation(s)
- Solène Barbeau
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33600 Pessac, France; (S.B.); (G.C.); (I.B.); (V.F.-M.); (C.G.); (R.M.); (P.B.); (P.V.); (J.-F.Q.)
- INSERM (Institut National de la Santé Et de la Recherche Médicale), Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, F-33600 Pessac, France
| | - Alexandre Joushomme
- Laboratoire de l’Intégration du Matériau au Système, UMR5518, Univ. Bordeaux, F-33400 Talence, France; (A.J.); (Y.C.); (Y.P.)
- CNRS (Centre National de la Recherche Scientifique), Laboratoire de L’integration du Matériau au Système, UMR5518, F-33400 Talence, France
| | - Yann Chappe
- Laboratoire de l’Intégration du Matériau au Système, UMR5518, Univ. Bordeaux, F-33400 Talence, France; (A.J.); (Y.C.); (Y.P.)
- CNRS (Centre National de la Recherche Scientifique), Laboratoire de L’integration du Matériau au Système, UMR5518, F-33400 Talence, France
| | - Guillaume Cardouat
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33600 Pessac, France; (S.B.); (G.C.); (I.B.); (V.F.-M.); (C.G.); (R.M.); (P.B.); (P.V.); (J.-F.Q.)
- INSERM (Institut National de la Santé Et de la Recherche Médicale), Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, F-33600 Pessac, France
| | - Isabelle Baudrimont
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33600 Pessac, France; (S.B.); (G.C.); (I.B.); (V.F.-M.); (C.G.); (R.M.); (P.B.); (P.V.); (J.-F.Q.)
- INSERM (Institut National de la Santé Et de la Recherche Médicale), Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, F-33600 Pessac, France
| | - Véronique Freund-Michel
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33600 Pessac, France; (S.B.); (G.C.); (I.B.); (V.F.-M.); (C.G.); (R.M.); (P.B.); (P.V.); (J.-F.Q.)
- INSERM (Institut National de la Santé Et de la Recherche Médicale), Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, F-33600 Pessac, France
| | - Christelle Guibert
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33600 Pessac, France; (S.B.); (G.C.); (I.B.); (V.F.-M.); (C.G.); (R.M.); (P.B.); (P.V.); (J.-F.Q.)
- INSERM (Institut National de la Santé Et de la Recherche Médicale), Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, F-33600 Pessac, France
| | - Roger Marthan
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33600 Pessac, France; (S.B.); (G.C.); (I.B.); (V.F.-M.); (C.G.); (R.M.); (P.B.); (P.V.); (J.-F.Q.)
- INSERM (Institut National de la Santé Et de la Recherche Médicale), Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, F-33600 Pessac, France
- CHU (Centre Hospitalier Universitaire) Bordeaux, Service d’Exploration Fonctionnelle Respiratoire, F-33600 Pessac, France
| | - Patrick Berger
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33600 Pessac, France; (S.B.); (G.C.); (I.B.); (V.F.-M.); (C.G.); (R.M.); (P.B.); (P.V.); (J.-F.Q.)
- INSERM (Institut National de la Santé Et de la Recherche Médicale), Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, F-33600 Pessac, France
- CHU (Centre Hospitalier Universitaire) Bordeaux, Service d’Exploration Fonctionnelle Respiratoire, F-33600 Pessac, France
| | - Pierre Vacher
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33600 Pessac, France; (S.B.); (G.C.); (I.B.); (V.F.-M.); (C.G.); (R.M.); (P.B.); (P.V.); (J.-F.Q.)
- INSERM (Institut National de la Santé Et de la Recherche Médicale), Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, F-33600 Pessac, France
| | - Yann Percherancier
- Laboratoire de l’Intégration du Matériau au Système, UMR5518, Univ. Bordeaux, F-33400 Talence, France; (A.J.); (Y.C.); (Y.P.)
- CNRS (Centre National de la Recherche Scientifique), Laboratoire de L’integration du Matériau au Système, UMR5518, F-33400 Talence, France
| | - Jean-François Quignard
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33600 Pessac, France; (S.B.); (G.C.); (I.B.); (V.F.-M.); (C.G.); (R.M.); (P.B.); (P.V.); (J.-F.Q.)
- INSERM (Institut National de la Santé Et de la Recherche Médicale), Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, F-33600 Pessac, France
| | - Thomas Ducret
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33600 Pessac, France; (S.B.); (G.C.); (I.B.); (V.F.-M.); (C.G.); (R.M.); (P.B.); (P.V.); (J.-F.Q.)
- INSERM (Institut National de la Santé Et de la Recherche Médicale), Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, F-33600 Pessac, France
| |
Collapse
|
92
|
Miao S, Zhou J, Liu B, Lei X, Wang T, Hao X, Cheng P, Wu H, Song Y, Pei G, Bi L. A 3D bioprinted nano-laponite hydrogel construct promotes osteogenesis by activating PI3K/AKT signaling pathway. Mater Today Bio 2022; 16:100342. [PMID: 35847377 PMCID: PMC9278073 DOI: 10.1016/j.mtbio.2022.100342] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 06/21/2022] [Accepted: 06/23/2022] [Indexed: 11/23/2022] Open
Abstract
Development of nano-laponite as bioinks based on cell-loaded hydrogels has recently attracted significant attention for promoting bone defect repairs and regeneration. However, the underlying mechanisms of the positive function of laponite in hydrogel was not fully explored. In this study, the effect of 3D bioprinted nano-laponite hydrogel construct on bone regeneration and the potential mechanism was explored in vitro and in vivo. In vitro analyses showed that the 3D construct protected encapsulated cells from shear stresses during bioprinting, promoted cell growth and cell spreading, and BMSCs at a density of 107/mL exhibited an optimal osteogenesis potential. Osteogenic differentiation and ectopic bone formation of BMSCs encapsulated inside the 3D construct were explored by determination of calcium deposition and x-ray, micro-CT analysis, respectively. RNA sequencing revealed that activation of PI3K/AKT signaling pathway of BMSCs inside the laponite hydrogel significantly upregulated expression of osteogenic related proteins. Expression of osteogenic proteins was significantly downregulated when the PI3K/AKT pathway was inhibited. The 3D bioprinted nano-laponite hydrogel construct exhibited a superior ability for bone regeneration in rat bones with defects compared with groups without laponite as shown by micro-CT and histological examination, while the osteogenesis activity was weakened by applications of a PI3K inhibitor. In summary, the 3D bioprinted nano-laponite hydrogel construct promoted bone osteogenesis by promoting cell proliferation, differentiation through activation of the PI3K/AKT signaling pathway.
Collapse
|
93
|
Wu S, He H, Huang J, Jiang S, Deng X, Huang J, Chen Y, Jiang Y. FMR1 is identified as an immune-related novel prognostic biomarker for renal clear cell carcinoma: A bioinformatics analysis of TAZ/YAP. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2022; 19:9295-9320. [PMID: 35942760 DOI: 10.3934/mbe.2022432] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
WW domain-containing transcription regulator 1 (TAZ, or WWTR1) and Yes-associated protein 1 (YAP) are both important effectors of the Hippo pathway and exhibit different functions. However, few studies have explored their co-regulatory mechanisms in kidney renal clear cell carcinoma (KIRC). Here, we used bioinformatics approaches to evaluate the co-regulatory roles of TAZ/YAP and screen novel biomarkers in KIRC. GSE121689 and GSE146354 were downloaded from the GEO. The limma was applied to identify the differential expression genes (DEGs) and the Venn diagram was utilized to screen co-expressed DEGs. Co-expressed DEGs obtained the corresponding pathways through GO and KEGG analysis. The protein-protein interaction (PPI) network was constructed using STRING. The hub genes were selected applying MCODE and CytoHubba. GSEA was further applied to identify the hub gene-related signaling pathways. The expression, survival, receiver operating character (ROC), and immune infiltration of the hub genes were analyzed by HPA, UALCAN, GEPIA, pROC, and TIMER. A total of 51 DEGs were co-expressed in the two datasets. The KEGG results showed that the enriched pathways were concentrated in the TGF-β signaling pathway and endocytosis. In the PPI network, the hub genes (STAU2, AGO2, FMR1) were identified by the MCODE and CytoHubba. The GSEA results revealed that the hub genes were correlated with the signaling pathways of metabolism and immunomodulation. We found that STAU2 and FMR1 were weakly expressed in tumors and were negatively associated with the tumor stages. The overall survival (OS) and disease-free survival (DFS) rate of the high-expressed group of FMR1 was greater than that of the low-expressed group. The ROC result exhibited that FMR1 had certainly a predictive ability. The TIMER results indicated that FMR1 was positively correlated to immune cell infiltration. The abovementioned results indicated that TAZ/YAP was involved in the TGF-β signaling pathway and endocytosis. FMR1 possibly served as an immune-related novel prognostic gene in KIRC.
Collapse
Affiliation(s)
- Sufang Wu
- The Key Laboratory of Model Animal and Stem Cell Biology in Hunan Province, Hunan Normal University, Changsha 410013, Hunan, China
- School of Medicine, Hunan Normal University, Changsha 410013, Hunan, China
| | - Hua He
- The Key Laboratory of Model Animal and Stem Cell Biology in Hunan Province, Hunan Normal University, Changsha 410013, Hunan, China
- School of Medicine, Hunan Normal University, Changsha 410013, Hunan, China
| | - Jingjing Huang
- The Key Laboratory of Model Animal and Stem Cell Biology in Hunan Province, Hunan Normal University, Changsha 410013, Hunan, China
- School of Medicine, Hunan Normal University, Changsha 410013, Hunan, China
| | - Shiyao Jiang
- The Key Laboratory of Model Animal and Stem Cell Biology in Hunan Province, Hunan Normal University, Changsha 410013, Hunan, China
- School of Medicine, Hunan Normal University, Changsha 410013, Hunan, China
| | - Xiyun Deng
- The Key Laboratory of Model Animal and Stem Cell Biology in Hunan Province, Hunan Normal University, Changsha 410013, Hunan, China
- School of Medicine, Hunan Normal University, Changsha 410013, Hunan, China
| | - Jun Huang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Yuanbing Chen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Yiqun Jiang
- The Key Laboratory of Model Animal and Stem Cell Biology in Hunan Province, Hunan Normal University, Changsha 410013, Hunan, China
- School of Medicine, Hunan Normal University, Changsha 410013, Hunan, China
| |
Collapse
|
94
|
Housmans BAC, Neefjes M, Surtel DAM, Vitík M, Cremers A, van Rhijn LW, van der Kraan PM, van den Akker GGH, Welting TJM. Synovial fluid from end-stage osteoarthritis induces proliferation and fibrosis of articular chondrocytes via MAPK and RhoGTPase signaling. Osteoarthritis Cartilage 2022; 30:862-874. [PMID: 35176481 DOI: 10.1016/j.joca.2021.12.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 02/02/2023]
Abstract
OBJECTIVES Alterations in the composition of synovial fluid have been associated with adverse effects on cartilage integrity and function. Here, we examined the phenotypic and proliferative behavior of human articular chondrocytes when cultured in vitro for 13 days with synovial fluid derived from end-stage osteoarthritis patients. MATERIALS AND METHODS Chondrocyte proliferation and phenotypical changes induced by osteoarthritic synovial fluid were analyzed using DNA staining, RT-qPCR, immunostainings, and immunoblotting. The molecular mechanisms by which osteoarthritic synovial fluid induced fibrosis and proliferation were studied using a phospho-protein antibody array and luciferase-based transcription factor activity assays. Specific pathway inhibitors were used to probe the involvement of pathways in fibrosis and proliferation. RESULTS Prolonged stimulation with osteoarthritic synovial fluid sustained chondrocyte proliferation and induced profound phenotypic changes, favoring a fibrotic over a chondrogenic or hypertrophic phenotype. A clear loss of chondrogenic markers at both the transcriptional and protein level was observed, while expression of several fibrosis-associated markers were upregulated over time. Phospho-kinase analysis revealed activation of MAPK and RhoGTPase signaling pathways by osteoarthritic synovial fluid, which was confirmed by elevated transcriptional activity of Elk-1 and SRF. Inhibitor studies revealed that ERK played a central role in the loss of chondrocyte phenotype, while EGFR and downstream mediators p38, JNK and Rac/Cdc42 were essential for fibrosis-associated collagen expression. Finally, we identified EGF signaling as a key activator of chondrocyte proliferation. CONCLUSIONS Osteoarthritic synovial fluid promoted chondrocyte fibrosis and proliferation through EGF receptor activation and downstream MAPK and RhoGTPase signaling.
Collapse
Affiliation(s)
- B A C Housmans
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, the Netherlands
| | - M Neefjes
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - D A M Surtel
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, the Netherlands
| | - M Vitík
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, the Netherlands
| | - A Cremers
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, the Netherlands
| | - L W van Rhijn
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - P M van der Kraan
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - G G H van den Akker
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, the Netherlands
| | - T J M Welting
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, the Netherlands; Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center+, Maastricht, the Netherlands.
| |
Collapse
|
95
|
Ravasio A, Morselli E, Bertocchi C. Mechanoautophagy: Synergies Between Autophagy and Cell Mechanotransduction at Adhesive Complexes. Front Cell Dev Biol 2022; 10:917662. [PMID: 35721483 PMCID: PMC9198486 DOI: 10.3389/fcell.2022.917662] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 05/13/2022] [Indexed: 12/15/2022] Open
Abstract
Cells are exposed and respond to various mechanical forces and physical cues stemming from their environment. This interaction has been seen to differentially regulate various cellular processes for maintenance of homeostasis, of which autophagy represents one of the major players. In addition, autophagy has been suggested to regulate mechanical functions of the cells including their interaction with the environment. In this minireview, we summarize the state of the art of the fascinating interplay between autophagy and the mechanotransduction machinery associated with cell adhesions, that we name ¨Mechanoautophagy¨
Collapse
Affiliation(s)
- Andrea Ravasio
- Institute for Biological and Medical Engineering Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
- *Correspondence: Cristina Bertocchi, ; Andrea Ravasio,
| | - Eugenia Morselli
- Department of Basic Sciences, Faculty of Medicine and Sciences, Universidad San Sebastián, Santiago, Chile
| | - Cristina Bertocchi
- Laboratory for Molecular Mechanics of Cell Adhesion, Department of Physiology Pontificia Universidad Católica de Chile, Santiago, Chile
- *Correspondence: Cristina Bertocchi, ; Andrea Ravasio,
| |
Collapse
|
96
|
Liu XY, Zhou K, Tian KJ, Yan BJ, Ren Z, Zhou ZX, Xiong WH, Jiang ZS. Hippo: a new hub for atherosclerotic disease. Curr Pharm Des 2022; 28:1321-1328. [DOI: 10.2174/1381612828666220428090540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 03/15/2022] [Indexed: 11/22/2022]
Abstract
Abstract:
Hippo,an evolutionarily conserved kinase cascade reaction in organisms,can respond to a set of signals,such as mechanical signals and cell metabolism,to maintain cell growth,differentiation,tissue/organ development and homeostasis.In the past ten years,HIPPO has controlled the development of tissues and organs by regulating the process of cell proliferation,especially in the field of cardiac regeneration after myocardial infarction.This suggests that HIPPO signaling is closely linked to cardiovascular disease.Atherosclerosis is the most common disease of the cardiovascular system. It is characterised by chronic inflammation of the vascular wall, mainly involving dysfunction of endothelial cells, smooth muscle cells and macrophages.Oxidized Low density lipoprotein (LDL) damages the barrier function of endothelial cells, which enter the middle membrane of the vascular wall, accelerates the formation of foam cells and promotes the occurrence and development of atherosclerosis.Autophagy is associated with the development of atherosclerosis.However, the mechanism of HIPPO regulation of atherosclerosis has not meant to clarified.In view of the pivotal role of this signaling pathway in maintaining cell growth,proliferation and differentiation,the imbalance of Hippo is related to atherosclerosis and related diseases.In this review,we emphasized Hippo as a hub for regulating atherosclerosis and discussed its potential targets in pathophysiology,human diseases,and related pharmacology.
Collapse
Affiliation(s)
- Xi-Yan Liu
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, China, 421001
| | - Kun Zhou
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, China, 421001
| | - Kai-Jiang Tian
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, China, 421001
| | - Bin-Jie Yan
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, China, 421001
| | - Zhong Ren
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, China, 421001
| | - Zhi-Xiang Zhou
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, China, 421001
| | - Wen-Hao Xiong
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, China, 421001
| | - Zhi-Sheng Jiang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, China, 421001
| |
Collapse
|
97
|
Regulator of G protein signaling 2 inhibits Gα q-dependent uveal melanoma cell growth. J Biol Chem 2022; 298:101955. [PMID: 35452684 PMCID: PMC9120238 DOI: 10.1016/j.jbc.2022.101955] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 04/06/2022] [Accepted: 04/07/2022] [Indexed: 12/23/2022] Open
Abstract
Activating mutations in Gαq/11 are a major driver of uveal melanoma (UM), the most common intraocular cancer in adults. While progress has recently been made in targeting Gαq/11 for UM therapy, the crucial role for these proteins in normal physiology and their high structural similarity with many other important GTPase proteins renders this approach challenging. The aim of the current study was to validate whether a key regulator of Gq signaling, regulator of G protein signaling 2 (RGS2), can inhibit Gαq-mediated UM cell growth. We used two UM cell lines, 92.1 and Mel-202, which both contain the most common activating mutation GαqQ209L and developed stable cell lines with doxycycline-inducible RGS2 protein expression. Using cell viability assays, we showed that RGS2 could inhibit cell growth in both of these UM cell lines. We also found that this effect was independent of the canonical GTPase-activating protein activity of RGS2 but was dependent on the association between RGS2 and Gαq. Furthermore, RGS2 induction resulted in only partial reduction in cell growth as compared to siRNA-mediated Gαq knockdown, perhaps because RGS2 was only able to reduce mitogen-activated protein kinase signaling downstream of phospholipase Cβ, while leaving activation of the Hippo signaling mediators yes-associated protein 1/TAZ, the other major pathway downstream of Gαq, unaffected. Taken together, our data indicate that RGS2 can inhibit UM cancer cell growth by associating with GαqQ209L as a partial effector antagonist.
Collapse
|
98
|
Caire R, Dalix E, Chafchafi M, Thomas M, Linossier MT, Normand M, Guignandon A, Vico L, Marotte H. YAP Transcriptional Activity Dictates Cell Response to TNF In Vitro. Front Immunol 2022; 13:856247. [PMID: 35401557 PMCID: PMC8989468 DOI: 10.3389/fimmu.2022.856247] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 02/23/2022] [Indexed: 01/15/2023] Open
Abstract
YAP/TAZ are transcription co-factors recently described responsive to pro-inflammatory cytokines and involved in inflammatory-related disorders. However, the role of tumor necrosis factor (TNF), a major pro-inflammatory cytokine, on YAP signaling is not well understood and controversial. Here, we observe in vitro, using wild type and YAP knockout HEK293 cells, that TNF triggers YAP nuclear translocation and transcriptional activity, thus being dependent on Rho family of GTPases. In response to TNF, YAP transcriptional activity orientates cell fate toward survival. Transcriptional analysis with Nanostring technology reveals that YAP modulates TNF-induced increase in fibro-inflammatory pathways such as NF-κB, inflammasomes, cytokines or chemokines signaling and pro-fibrotic pathways involving TGF-β and extracellular matrix remodeling. Therefore, in response to TNF, YAP acts as a sustainer of the inflammatory response and as a molecular link between inflammation and fibrotic processes. This work identifies that YAP is critical to drive several biological effects of TNF which are involved in cancer and inflammatory disorders.
Collapse
Affiliation(s)
- Robin Caire
- INSERM, U1059-SAINBIOSE, Université de Lyon, Saint-Etienne, France
| | - Elisa Dalix
- INSERM, U1059-SAINBIOSE, Université de Lyon, Saint-Etienne, France
| | - Marwa Chafchafi
- INSERM, U1059-SAINBIOSE, Université de Lyon, Saint-Etienne, France
| | - Mireille Thomas
- INSERM, U1059-SAINBIOSE, Université de Lyon, Saint-Etienne, France
| | | | - Myriam Normand
- INSERM, U1059-SAINBIOSE, Université de Lyon, Saint-Etienne, France
| | - Alain Guignandon
- INSERM, U1059-SAINBIOSE, Université de Lyon, Saint-Etienne, France
| | - Laurence Vico
- INSERM, U1059-SAINBIOSE, Université de Lyon, Saint-Etienne, France
| | - Hubert Marotte
- INSERM, U1059-SAINBIOSE, Université de Lyon, Saint-Etienne, France.,Department of Rheumatology, Hôpital Nord, University Hospital Saint-Etienne, Saint-Etienne, France
| |
Collapse
|
99
|
Zhang W, Zhang Y, Li X, Cao Z, Mo Q, Sheng R, Ling C, Chi J, Yao Q, Chen J, Wang H. Multifunctional polyphenol-based silk hydrogel alleviates oxidative stress and enhances endogenous regeneration of osteochondral defects. Mater Today Bio 2022; 14:100251. [PMID: 35469254 PMCID: PMC9034395 DOI: 10.1016/j.mtbio.2022.100251] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 03/08/2022] [Accepted: 03/31/2022] [Indexed: 01/25/2023] Open
Abstract
In osteochondral defects, oxidative stress caused by elevated levels of reactive oxygen species (ROS) can disrupt the normal endogenous repair process. In this study, a multifunctional hydrogel composed of silk fibroin (SF) and tannic acid (TA), the FDA-approved ingredients, was developed to alleviate oxidative stress and enhance osteochondral regeneration. In this proposed hydrogel, SF first interacts with TA to form a hydrogen-bonded supramolecular structure, which is subsequently enzymatically crosslinked to form a stable hydrogel. Furthermore, TA had multiple phenolic hydroxyl groups that formed interactions with the therapeutic molecule E7 peptide for controlled drug delivery. In vitro investigations showed that SF-TA and SF-TA-E7 hydrogels exhibited a multitude of biological effects including scavenging of ROS, maintaining cell viability, and promoting the proliferation of bone marrow mesenchymal stem cells (BMSCs) against oxidative stress. The proteomic analysis indicated that SF-TA and SF-TA-E7 hydrogels suppressed oxidative stress, which in turn improved cell proliferation in multiple proliferation and apoptosis-related pathways. In rabbit osteochondral defect model, SF-TA and SF-TA-E7 hydrogels promoted enhanced regeneration of both cartilage and subchondral bone as compared to hydrogel without TA incorporation. These findings indicated that the multifunctional SF-TA hydrogel provided a microenvironment suitable for the endogenous regeneration of osteochondral defects.
Collapse
Affiliation(s)
- Wei Zhang
- School of Medicine, Southeast University, 210009, Nanjing, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210096, Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), China
- Corresponding author. School of Medicine, Southeast University, 210009, Nanjing, China.
| | - Yanan Zhang
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Xiaolong Li
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Zhicheng Cao
- School of Medicine, Southeast University, 210009, Nanjing, China
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006, Nanjing, China
| | - Qingyun Mo
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Renwang Sheng
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Chen Ling
- School of Medicine, Southeast University, 210009, Nanjing, China
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006, Nanjing, China
| | - Jiayu Chi
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Qingqiang Yao
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006, Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), China
- Corresponding author. Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006, Nanjing, China.
| | - Jialin Chen
- School of Medicine, Southeast University, 210009, Nanjing, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210096, Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), China
- Corresponding author. School of Medicine, Southeast University, 210009, Nanjing, China.
| | - Hongmei Wang
- School of Medicine, Southeast University, 210009, Nanjing, China
- Department of Pharmaceutical Sciences, Binzhou Medical University, 264003, Yantai, Shandong, China
- Corresponding author. School of Medicine, Southeast University, 210009, Nanjing, China.
| |
Collapse
|
100
|
Astolfi M, Rispoli G, Benedusi M, Zonta G, Landini N, Valacchi G, Malagù C. Chemoresistive Sensors for Cellular Type Discrimination Based on Their Exhalations. NANOMATERIALS 2022; 12:nano12071111. [PMID: 35407231 PMCID: PMC9000844 DOI: 10.3390/nano12071111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 03/24/2022] [Accepted: 03/25/2022] [Indexed: 12/03/2022]
Abstract
The detection of volatile organic compounds (VOCs) exhaled by human body fluids is a recent and promising method to reveal tumor formations. In this feasibility study, a patented device, based on nanostructured chemoresistive gas sensors, was employed to explore the gaseous exhalations of tumoral, immortalized, and healthy cell lines, with the aim of distinguishing their VOC patterns. The analysis of the device output to the cell VOCs, emanated at different incubation times and initial plating concentrations, was performed to evaluate the device suitability to identify the cell types and to monitor their growth. The sensors ST25 (based on tin and titanium oxides), STN (based on tin, titanium, and niobium oxides), and TiTaV (based on titanium, tantalum and vanadium oxides) used here, gave progressively increasing responses upon the cell density increase and incubation time; the sensor W11 (based on tungsten oxide) gave instead unreliable responses to all cell lines. All sensors (except for W11) gave large and consistent responses to RKO and HEK293 cells, while they were less responsive to CHO, A549, and CACO-2 ones. The encouraging results presented here, although preliminary, foresee the development of sensor arrays capable of identifying tumor presence and its type.
Collapse
Affiliation(s)
- Michele Astolfi
- Department of Physics and Earth Sciences, University of Ferrara, 44122 Ferrara, Italy; (G.Z.); (N.L.); (C.M.)
- SCENT S.r.l., Via Quadrifoglio 11, 44124 Ferrara, Italy
- Correspondence: (M.A.); (G.R.)
| | - Giorgio Rispoli
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy;
- Correspondence: (M.A.); (G.R.)
| | - Mascia Benedusi
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy;
| | - Giulia Zonta
- Department of Physics and Earth Sciences, University of Ferrara, 44122 Ferrara, Italy; (G.Z.); (N.L.); (C.M.)
- SCENT S.r.l., Via Quadrifoglio 11, 44124 Ferrara, Italy
| | - Nicolò Landini
- Department of Physics and Earth Sciences, University of Ferrara, 44122 Ferrara, Italy; (G.Z.); (N.L.); (C.M.)
- SCENT S.r.l., Via Quadrifoglio 11, 44124 Ferrara, Italy
| | - Giuseppe Valacchi
- Department of Environmental Science and Prevention, University of Ferrara, 44121 Ferrara, Italy;
- Plants for Human Health Institute, NC State University, Kannapolis, NC 28081, USA
- Department of Food and Nutrition, Kyung Hee University, Seoul 130-701, Korea
| | - Cesare Malagù
- Department of Physics and Earth Sciences, University of Ferrara, 44122 Ferrara, Italy; (G.Z.); (N.L.); (C.M.)
- SCENT S.r.l., Via Quadrifoglio 11, 44124 Ferrara, Italy
| |
Collapse
|