1
|
Vasu M, Ahlawat S, Arora R, Sharma R. Deciphering the molecular drivers for cashmere/pashmina fiber production in goats: a comprehensive review. Mamm Genome 2025; 36:162-182. [PMID: 39904908 DOI: 10.1007/s00335-025-10109-z] [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: 11/29/2024] [Accepted: 01/29/2025] [Indexed: 02/06/2025]
Abstract
Cashmere, also known as pashmina, is derived from the secondary hair follicles of Cashmere/Changthangi goats. Renowned as the world's most luxurious natural fiber, it holds significant economic value in the textile industry. This comprehensive review enhances our understanding of the complex biological processes governing cashmere/pashmina fiber development and quality, enabling advancements in selective breeding and fiber enhancement strategies. The review specifically examines the molecular determinants influencing fiber development, with an emphasis on keratins (KRTs) and keratin-associated proteins (KRTAPs). It also explores the roles of key molecular pathways, including Wnt, Notch, BMP, NF-kappa B, VEGF, cAMP, PI3K-Akt, ECM, cell adhesion, Hedgehog, MAPK, Ras, JAK-STAT, TGF-β, mTOR, melanogenesis, FoxO, Hippo, and Rap1 signaling. Understanding these intricate molecular cascades provides valuable insights into the mechanisms orchestrating hair follicle growth, further advancing the biology of this coveted natural fiber. Expanding multi-omics approaches will enhance breeding precision and deepen our understanding of molecular pathways influencing cashmere production. Future research should address critical gaps, such as the impact of environmental factors, epigenetic modifications, and functional studies of genetic variants. Collaboration among breeders, researchers, and policymakers is essential for translating genomic advancements into practical applications. Such efforts can promote sustainable practices, conserve biodiversity, and ensure the long-term viability of high-quality cashmere production. Aligning genetic insights with conservation strategies will support the sustainable growth of the cashmere industry while preserving its economic and ecological value.
Collapse
Affiliation(s)
- Mahanthi Vasu
- ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana, India
| | - Sonika Ahlawat
- ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana, India.
| | - Reena Arora
- ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana, India
| | - Rekha Sharma
- ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana, India
| |
Collapse
|
2
|
Yousefi F, Foster LA, Selim OA, Zhao C. Integrating Physical and Biochemical Cues for Muscle Engineering: Scaffolds and Graft Durability. Bioengineering (Basel) 2024; 11:1245. [PMID: 39768063 PMCID: PMC11673930 DOI: 10.3390/bioengineering11121245] [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: 10/23/2024] [Revised: 11/26/2024] [Accepted: 11/28/2024] [Indexed: 01/11/2025] Open
Abstract
Muscle stem cells (MuSCs) are essential for skeletal muscle regeneration, influenced by a complex interplay of mechanical, biochemical, and molecular cues. Properties of the extracellular matrix (ECM) such as stiffness and alignment guide stem cell fate through mechanosensitive pathways, where forces like shear stress translate into biochemical signals, affecting cell behavior. Aging introduces senescence which disrupts the MuSC niche, leading to reduced regenerative capacity via epigenetic alterations and metabolic shifts. Transplantation further challenges MuSC viability, often resulting in fibrosis driven by dysregulated fibro-adipogenic progenitors (FAPs). Addressing these issues, scaffold designs integrated with pharmacotherapy emulate ECM environments, providing cues that enhance graft functionality and endurance. These scaffolds facilitate the synergy between mechanotransduction and intracellular signaling, optimizing MuSC proliferation and differentiation. Innovations utilizing human pluripotent stem cell-derived myogenic progenitors and exosome-mediated delivery exploit bioactive properties for targeted repair. Additionally, 3D-printed and electrospun scaffolds with adjustable biomechanical traits tackle scalability in treating volumetric muscle loss. Advanced techniques like single-cell RNA sequencing and high-resolution imaging unravel muscle repair mechanisms, offering precise mapping of cellular interactions. Collectively, this interdisciplinary approach fortifies tissue graft durability and MuSC maintenance, propelling therapeutic strategies for muscle injuries and degenerative diseases.
Collapse
Affiliation(s)
- Farbod Yousefi
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (F.Y.); (L.A.F.); (O.A.S.)
| | - Lauren Ann Foster
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (F.Y.); (L.A.F.); (O.A.S.)
- Atlanta Veterans Affairs Medical Center, Emory University School of Medicine, Atlanta, GA 30307, USA
| | - Omar A. Selim
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (F.Y.); (L.A.F.); (O.A.S.)
| | - Chunfeng Zhao
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (F.Y.); (L.A.F.); (O.A.S.)
| |
Collapse
|
3
|
Wang J, An Z, Wu Z, Zhou W, Sun P, Wu P, Dang S, Xue R, Bai X, Du Y, Chen R, Wang W, Huang P, Lam SM, Ai Y, Liu S, Shui G, Zhang Z, Liu Z, Huang J, Fang X, He K. Spatial organization of PI3K-PI(3,4,5)P 3-AKT signaling by focal adhesions. Mol Cell 2024; 84:4401-4418.e9. [PMID: 39488211 DOI: 10.1016/j.molcel.2024.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 08/17/2024] [Accepted: 10/08/2024] [Indexed: 11/04/2024]
Abstract
The class I phosphatidylinositol 3-kinase (PI3K)-AKT signaling pathway is a key regulator of cell survival, growth, and proliferation and is among the most frequently mutated pathways in cancer. However, where and how PI3K-AKT signaling is spatially activated and organized in mammalian cells remains poorly understood. Here, we identify focal adhesions (FAs) as subcellular signaling hubs organizing the activation of PI3K-PI(3,4,5)P3-AKT signaling in human cancer cells containing p110α mutations under basal conditions. We find that class IA PI3Ks are preferentially recruited to FAs for activation, resulting in localized production of PI(3,4,5)P3 around FAs. As the effector protein of PI(3,4,5)P3, AKT1 molecules are dynamically recruited around FAs for activation. The spatial recruitment/activation of the PI3K-PI(3,4,5)P3-AKT cascade is regulated by activated FA kinase (FAK). Furthermore, combined inhibition of p110α and FAK results in a more potent inhibitory effect on cancer cells. Thus, our results unveil a growth-factor independent, compartmentalized organization mechanism for PI3K-PI(3,4,5)P3-AKT signaling.
Collapse
Affiliation(s)
- Jing Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhengyang An
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhongsheng Wu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wei Zhou
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Pengyu Sun
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Piyu Wu
- The Institute for Advanced Studies, Wuhan University, Wuhan 430072, China
| | - Song Dang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Rui Xue
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China
| | - Xue Bai
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
| | - Yongtao Du
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Rongmei Chen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wenxu Wang
- The Institute for Advanced Studies, Wuhan University, Wuhan 430072, China
| | - Pei Huang
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Sin Man Lam
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; Lipidall Technologies Company Limited, Changzhou, Jiangsu 213000, China
| | - Youwei Ai
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Suling Liu
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai 200032, China
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhe Zhang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China; Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Zheng Liu
- The Institute for Advanced Studies, Wuhan University, Wuhan 430072, China
| | - Jianyong Huang
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China
| | - Xiaohong Fang
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.
| | - Kangmin He
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
4
|
Lou H, Lu H, Zhang S, Shi Y, Xu E, Liu D, Chen Q. Highly aligned myotubes formation of piscine satellite cells in 3D fibrin hydrogels of cultured meat. Int J Biol Macromol 2024; 282:136879. [PMID: 39490877 DOI: 10.1016/j.ijbiomac.2024.136879] [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: 08/13/2024] [Revised: 10/14/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024]
Abstract
Currently, various cultured meat products, including chicken, beef and pork, have been developed. However, established methods for production cultured fish meat with highly aligned myotubes are still lack. In this study, we introduced a culture method based on high-biocompatibility fibrin hydrogels with an easy-to-use tissue mold for obtaining cultured fish fillets that closely mimicked the structure of natural fish fillets. Results showed that highly aligned myotubes were observed within the muscle bundles culturing in the tissue mold. The myotube fusion index was also increased to 72.65 %. Furthermore, key differentiation genes (desmin, myosin light chain kinase, myocilin) were up-regulated in the tissue mold group. Transcriptomic analysis further supported the effectiveness of method in promoting myoblast fusion. Stiffness of the muscle bundles was also positively impacted by the tissue mold. Ultimately, sensory and nutritional characteristics of natural and cultured fish fillets were compared, revealing that cultured fish fillets prepared from the tissue mold was closer to natural fish fillets in sensory characteristic, and there were still some gaps with natural fish fillets in nutritional characteristic. Overall, our findings suggest that optimizing culture methods can help bridge some gaps between natural meat and cultured meat, facilitating the development of cultured fish meat.
Collapse
Affiliation(s)
- Hanghang Lou
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310058, China
| | - Hongyun Lu
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310058, China
| | - Shengliang Zhang
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310058, China
| | - Ying Shi
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310058, China
| | - Enbo Xu
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310058, China; Future Food Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China
| | - Donghong Liu
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310058, China; Future Food Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China
| | - Qihe Chen
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310058, China; Future Food Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China.
| |
Collapse
|
5
|
Kliewe F, Siegerist F, Hammer E, Al-Hasani J, Amling TRJ, Hollemann JZE, Schindler M, Drenic V, Simm S, Amann K, Daniel C, Lindenmeyer M, Hecker M, Völker U, Endlich N. Zyxin is important for the stability and function of podocytes, especially during mechanical stretch. Commun Biol 2024; 7:446. [PMID: 38605154 PMCID: PMC11009394 DOI: 10.1038/s42003-024-06125-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 03/29/2024] [Indexed: 04/13/2024] Open
Abstract
Podocyte detachment due to mechanical stress is a common issue in hypertension-induced kidney disease. This study highlights the role of zyxin for podocyte stability and function. We have found that zyxin is significantly up-regulated in podocytes after mechanical stretch and relocalizes from focal adhesions to actin filaments. In zyxin knockout podocytes, we found that the loss of zyxin reduced the expression of vinculin and VASP as well as the expression of matrix proteins, such as fibronectin. This suggests that zyxin is a central player in the translation of mechanical forces in podocytes. In vivo, zyxin is highly up-regulated in patients suffering from diabetic nephropathy and in hypertensive DOCA-salt treated mice. Furthermore, zyxin loss in mice resulted in proteinuria and effacement of podocyte foot processes that was measured by super resolution microscopy. This highlights the essential role of zyxin for podocyte maintenance in vitro and in vivo, especially under mechanical stretch.
Collapse
Affiliation(s)
- Felix Kliewe
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany.
| | - Florian Siegerist
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Elke Hammer
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Jaafar Al-Hasani
- Department of Cardiovascular Physiology, Heidelberg University, Heidelberg, Germany
| | | | | | - Maximilian Schindler
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Vedran Drenic
- NIPOKA GmbH, Center of High-End Imaging, Greifswald, Germany
| | - Stefan Simm
- Institute of Bioinformatics, University Medicine Greifswald, Greifswald, Germany
| | - Kerstin Amann
- Department of Nephropathology; Friedrich-Alexander University (FAU) Erlangen-Nuremberg, Erlangen, Germany
| | - Christoph Daniel
- Department of Nephropathology; Friedrich-Alexander University (FAU) Erlangen-Nuremberg, Erlangen, Germany
| | - Maja Lindenmeyer
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Markus Hecker
- Department of Cardiovascular Physiology, Heidelberg University, Heidelberg, Germany
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Nicole Endlich
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
- NIPOKA GmbH, Center of High-End Imaging, Greifswald, Germany
| |
Collapse
|
6
|
Pan G, Xie H, Xia Y. Disulfidptosis characterizes the tumor microenvironment and predicts immunotherapy sensitivity and prognosis in bladder cancer. Heliyon 2024; 10:e25573. [PMID: 38356551 PMCID: PMC10864973 DOI: 10.1016/j.heliyon.2024.e25573] [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: 07/28/2023] [Revised: 01/13/2024] [Accepted: 01/29/2024] [Indexed: 02/16/2024] Open
Abstract
Background Bladder cancer (BLCA) is prone to metastasis and has poor prognosis with unsatisfactory treatment responsiveness. Disulfidptosis is a recently discovered, novel mode of cell death that is closely associated with human cancers. However, a comprehensive analysis of the relationship between disulfidptosis and BLCA is lacking. Therefore, this study aimed to explore the potential effect of disulfidptosis on BLCA and identify a biomarker for evaluating the prognosis and immunotherapy of patients with BLCA. Material and methods We acquired BLCA RNA sequencing data from The Cancer Genome Atlas Urothelial Bladder Carcinoma (TCGA-BLCA) cohort (containing 19 normal samples and 409 tumor samples) and the GES39281 cohort (containing 94 tumor samples) which were used for external validation of the signature. Initially, we performed unsupervised consensus clustering to explore disulfidptosis-related subgroups. We then conducted functional enrichment analysis on these subgroups to gain insights into their biological significance and evaluate their immunotherapy response and chemotherapy sensitivity. Next, we conducted Least Absolute Shrinkage and Selection Operator (LASSO) regression and multivariate Cox regression to construct a prognostic signature in the TCGA training set for prognosis-related differentially expressed genes (DEGs) in the disulfidptosis-related subgroups. Subsequently, we used a receiver operating characteristic (ROC) curve and independent prognostic analysis to validate the predictive performance of the signature in the TCGA testing and the GES39281 cohorts. Finally, we explored the therapeutic value of this signature in patients with BLCA, in terms of immunotherapy and chemotherapy. Result In this study, we obtained two subgroups: DRG-high (238 samples) and DRG-low (160 samples). The DRG-high group exhibited a poor survival rate compared to the DRG-low group and had a significant association with tumor grade, stage, and metastasis. Additionally, several pathways related to cancer and the immune system were enriched in the high-DRG group. Moreover, the DRG-high group exhibited higher expression of PD1 and CTLA4 and had a better response to immunotherapy in patients with both PD1 and CTLA4 positivity. Conversely, the DRG-high group was more sensitive to common chemotherapeutic agents. A prognostic signature was created, consisting of COL5A1, DIRAS3, NKG7, and POLR3G and validated as having a robust predictive capability. Patients in the low-risk-score group had more immune cells associated with tumor suppression and better immunotherapy outcomes. Conclusion This study contributes to our understanding of the characteristics of disulfidptosis-related subgroups in BLCA. Disulfidptosis-related signatures can be used to assess the prognosis and immunotherapy of patients with BLCA.
Collapse
Affiliation(s)
- Guizhen Pan
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Huan Xie
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Yeye Xia
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Oncology, Chengdu Fifth People's Hospital, Sichuan, China
| |
Collapse
|
7
|
Cheng D, Wang J, Yao M, Cox CD. Joining forces: crosstalk between mechanosensitive PIEZO1 ion channels and integrin-mediated focal adhesions. Biochem Soc Trans 2023; 51:1897-1906. [PMID: 37772664 DOI: 10.1042/bst20230042] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 09/18/2023] [Accepted: 09/20/2023] [Indexed: 09/30/2023]
Abstract
Both integrin-mediated focal adhesions (FAs) and mechanosensitive ion channels such as PIEZO1 are critical in mechanotransduction processes that influence cell differentiation, development, and cancer. Ample evidence now exists for regulatory crosstalk between FAs and PIEZO1 channels with the molecular mechanisms underlying this process remaining unclear. However, an emerging picture is developing based on spatial crosstalk between FAs and PIEZO1 revealing a synergistic model involving the cytoskeleton, extracellular matrix (ECM) and calcium-dependent signaling. Already cell type, cell contractility, integrin subtypes and ECM composition have been shown to regulate this crosstalk, implying a highly fine-tuned relationship between these two major mechanosensing systems. In this review, we summarize the latest advances in this area, highlight the physiological implications of this crosstalk and identify gaps in our knowledge that will improve our understanding of cellular mechanosensing.
Collapse
Affiliation(s)
- Delfine Cheng
- The Victor Chang Cardiac Research Institute, Sydney, NSW 2010, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Kensington, NSW 2052, Australia
| | - Junfan Wang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen 518055, China
| | - Mingxi Yao
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen 518055, China
| | - Charles D Cox
- The Victor Chang Cardiac Research Institute, Sydney, NSW 2010, Australia
- School of Biomedical Sciences, Faculty of Medicine & Health, University of New South Wales, Kensington, NSW 2052, Australia
| |
Collapse
|
8
|
Wu MC, Yu HW, Chen YQ, Ou MH, Serrano R, Huang GL, Wang YK, Lin KH, Fan YJ, Wu CC, Del Álamo JC, Chiou A, Chien S, Kuo JC. Early committed polarization of intracellular tension in response to cell shape determines the osteogenic differentiation of mesenchymal stromal cells. Acta Biomater 2023; 163:287-301. [PMID: 36328121 PMCID: PMC11389728 DOI: 10.1016/j.actbio.2022.10.052] [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: 04/15/2022] [Revised: 10/24/2022] [Accepted: 10/25/2022] [Indexed: 11/07/2022]
Abstract
Within the heterogeneous tissue architecture, a comprehensive understanding of how cell shapes regulate cytoskeletal mechanics by adjusting focal adhesions (FAs) signals to correlate with the lineage commitment of mesenchymal stromal cells (MSCs) remains obscure. Here, via engineered extracellular matrices, we observed that the development of mature FAs, coupled with a symmetrical pattern of radial fiber bundles, appeared at the right-angle vertices in cells with square shape. While circular cells aligned the transverse fibers parallel to the cell edge, and moved them centripetally in a counter-clockwise direction, symmetrical bundles of radial fibers at the vertices of square cells disrupted the counter-clockwise swirling and bridged the transverse fibers to move centripetally. In square cells, the contractile force, generated by the myosin IIA-enriched transverse fibers, were concentrated and transmitted outwards along the symmetrical bundles of radial fibers, to the extracellular matrix through FAs, and thereby driving FA organization and maturation. The symmetrical radial fiber bundles concentrated the transverse fibers contractility inward to the linkage between the actin cytoskeleton and the nuclear envelope. The tauter cytoskeletal network adjusted the nuclear-actomyosin force balance to cause nuclear deformability and to increase nuclear translocation of the transcription co-activator YAP, which in turn modulated the switch in MSC commitment. Thus, FAs dynamically respond to geometric cues and remodel actin cytoskeletal network to re-distribute intracelluar tension towards the cell nucleus, and thereby controlling YAP mechanotransduction signaling in regulating MSC fate decision. STATEMENT OF SIGNIFICANCE: We decipher how cellular mechanics is self-organized depending on extracellular geometric features to correlate with mesenchymal stromal cell lineage commitment. In response to geometry constrains on cell morphology, symmetrical radial fiber bundles are assembled and clustered depending on the maturation state of focal adhesions and bridge with the transverse fibers, and thereby establishing the dynamic cytoskeletal network. Contractile force, generated by the myosin-IIA-enriched transverse fibers, is transmitted and dynamically drives the retrograde movement of the actin cytoskeletal network, which appropriately adjusts the nuclear-actomyosin force balance and deforms the cell nucleus for YAP mechano-transduction signaling in regulating mesenchymal stromal cell fate decision.
Collapse
Affiliation(s)
- Ming-Chung Wu
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Helen Wenshin Yu
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Yin-Quan Chen
- Cancer Progression Research Center, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Meng-Hsin Ou
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Ricardo Serrano
- Department of Bioengineering and Institute of Engineering in Medicine, University of California at San Diego, La Jolla, CA 92093, USA; Department of Mechanical and Aerospace Engineering, University of California at San Diego, La Jolla, CA 92093, USA; Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Guan-Lin Huang
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Yang-Kao Wang
- Department of Cell Biology and Anatomy, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Kung-Hui Lin
- Institute of Physics, Academia Sinica, Taipei 11529, Taiwan
| | - Yu-Jui Fan
- School of Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan
| | - Chi-Chang Wu
- Department of Electronic Engineering, National Chin-Yi University of Technology, Taichung 411030, Taiwan
| | - Juan C Del Álamo
- Department of Bioengineering and Institute of Engineering in Medicine, University of California at San Diego, La Jolla, CA 92093, USA; Department of Mechanical and Aerospace Engineering, University of California at San Diego, La Jolla, CA 92093, USA; Center for Cardiovascular Biology, University of Washington, School of Medicine, Seattle, WA, 98109, USA; Mechanical Engineering Department, University of Washington, Seattle, WA, 98195, USA
| | - Arthur Chiou
- Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Shu Chien
- Department of Bioengineering and Institute of Engineering in Medicine, University of California at San Diego, La Jolla, CA 92093, USA
| | - Jean-Cheng Kuo
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; Cancer Progression Research Center, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan.
| |
Collapse
|
9
|
Kemper L, Hensel A. Campylobacter jejuni: targeting host cells, adhesion, invasion, and survival. Appl Microbiol Biotechnol 2023; 107:2725-2754. [PMID: 36941439 PMCID: PMC10027602 DOI: 10.1007/s00253-023-12456-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 02/21/2023] [Accepted: 02/23/2023] [Indexed: 03/23/2023]
Abstract
Campylobacter jejuni, causing strong enteritis, is an unusual bacterium with numerous peculiarities. Chemotactically controlled motility in viscous milieu allows targeted navigation to intestinal mucus and colonization. By phase variation, quorum sensing, extensive O-and N-glycosylation and use of the flagellum as type-3-secretion system C. jejuni adapts effectively to environmental conditions. C. jejuni utilizes proteases to open cell-cell junctions and subsequently transmigrates paracellularly. Fibronectin at the basolateral side of polarized epithelial cells serves as binding site for adhesins CadF and FlpA, leading to intracellular signaling, which again triggers membrane ruffling and reduced host cell migration by focal adhesion. Cell contacts of C. jejuni results in its secretion of invasion antigens, which induce membrane ruffling by paxillin-independent pathway. In addition to fibronectin-binding proteins, other adhesins with other target structures and lectins and their corresponding sugar structures are involved in host-pathogen interaction. Invasion into the intestinal epithelial cell depends on host cell structures. Fibronectin, clathrin, and dynein influence cytoskeletal restructuring, endocytosis, and vesicular transport, through different mechanisms. C. jejuni can persist over a 72-h period in the cell. Campylobacter-containing vacuoles, avoid fusion with lysosomes and enter the perinuclear space via dynein, inducing signaling pathways. Secretion of cytolethal distending toxin directs the cell into programmed cell death, including the pyroptotic release of proinflammatory substances from the destroyed cell compartments. The immune system reacts with an inflammatory cascade by participation of numerous immune cells. The development of autoantibodies, directed not only against lipooligosaccharides, but also against endogenous gangliosides, triggers autoimmune diseases. Lesions of the epithelium result in loss of electrolytes, water, and blood, leading to diarrhea, which flushes out mucus containing C. jejuni. Together with the response of the immune system, this limits infection time. Based on the structural interactions between host cell and bacterium, the numerous virulence mechanisms, signaling, and effects that characterize the infection process of C. jejuni, a wide variety of targets for attenuation of the pathogen can be characterized. The review summarizes strategies of C. jejuni for host-pathogen interaction and should stimulate innovative research towards improved definition of targets for future drug development. KEY POINTS: • Bacterial adhesion of Campylobacter to host cells and invasion into host cells are strictly coordinated processes, which can serve as targets to prevent infection. • Reaction and signalling of host cell depend on the cell type. • Campylobacter virulence factors can be used as targets for development of antivirulence drug compounds.
Collapse
Affiliation(s)
- Leon Kemper
- Institute of Pharmaceutical Biology and Phytochemistry, University of Münster, Corrensstraße 48, 48149, Münster, Germany
| | - Andreas Hensel
- Institute of Pharmaceutical Biology and Phytochemistry, University of Münster, Corrensstraße 48, 48149, Münster, Germany.
| |
Collapse
|
10
|
Wang X, Zhao N, Wang T, Du S, Liu Q, Li J. Transcriptome Analysis Provides Insights into Copulation, Fertilization, and Gestation in Sebastes schlegelii. Genes (Basel) 2022; 13:genes13101812. [PMID: 36292697 PMCID: PMC9601582 DOI: 10.3390/genes13101812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/26/2022] [Accepted: 09/29/2022] [Indexed: 11/16/2022] Open
Abstract
Among the viviparous marine teleosts of China, the black rockfish (Sebastes schlegelii Hilgendorf) is one of the most economically important. In addition to copulation and internal fertilization, it features lengthy sperm storage in the female ovary as well as a high rate of abortion. A network of gene regulation is necessary for these processes. To elucidate the mechanisms of copulation, fertilization, and gestation, it is essential to determine the genetic basis of viviparous teleost oogenesis and embryogenesis. In this study, we analyzed the transcriptome of the ovary during different developmental phases to investigate the dynamic changes that occur. We constructed 24 ovary transcriptomes. In order to investigate the regulation of embryogenesis, differentially expressed genes (DEGs) with specific expression patterns were subjected to gene ontology annotation, pathway analyses, and weighted gene co-expression network analysis (WGCNA). The up-regulated genes were significantly enriched in focal adhesion, regulation of the actin cytoskeleton, Wnt, and ECM-receptor interaction signaling pathways. As a result of our study, we provide omics evidence for copulation, fertilization, and gestation in viviparous marine teleosts. Decoding the S. schlegelii gene regulation network, as well as providing new insights into embryogenesis, is highly valuable to researchers in the marine teleost reproduction sciences.
Collapse
Affiliation(s)
- Xueying Wang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, China
| | - Ning Zhao
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tao Wang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, China
- Qingdao Agricultural University, Qingdao 266237, China
| | - Shuran Du
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, China
- Qingdao Agricultural University, Qingdao 266237, China
| | - Qinghua Liu
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, China
| | - Jun Li
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, China
- Correspondence: ; Tel./Fax: +86-532-82898718
| |
Collapse
|
11
|
The explorations of dynamic interactions of paxillin at the focal adhesions. BIOCHIMICA ET BIOPHYSICA ACTA. PROTEINS AND PROTEOMICS 2022; 1870:140825. [PMID: 35926716 DOI: 10.1016/j.bbapap.2022.140825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 07/16/2022] [Accepted: 07/27/2022] [Indexed: 11/20/2022]
Abstract
Paxillin is one of the most important adapters in integrin-mediated adhesions that performs numerous crucial functions relying on its dynamic interactions. Its structural behavior serves different purposes, providing a base for several activities. The various domains of paxillin display different functions in the whole process of cell movements and have a significant role in cell adhesion, migration, signal transmission, and protein-protein interactions. On the other hand, some paxillin-associated proteins provide a unique spatiotemporal mechanism for regulating its dynamic characteristics in the tissue homeostasis and make it a more complex and decisive protein at the focal adhesions. This review briefly describes the structural adaptations and molecular mechanisms of recruitment of paxillin into adhesions, explains paxillin's binding dynamics and impact on adhesion stability and turnover, and reveals a variety of paxillin-associated regulatory mechanisms and how paxillin is embedded into the signaling networks.
Collapse
|
12
|
Tuftelin 1 Facilitates Hepatocellular Carcinoma Progression through Regulation of Lipogenesis and Focal Adhesion Maturation. J Immunol Res 2022; 2022:1590717. [PMID: 35769513 PMCID: PMC9234046 DOI: 10.1155/2022/1590717] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 06/02/2022] [Indexed: 12/23/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common type of primary liver malignancy with poor prognosis worldwide. Emerging evidences demonstrated critical roles of lipid de novo synthesis in HCC progression, yet its regulatory mechanisms are not fully understood. Herein, we found that tuftelin 1 (TUFT1), an acidic phosphorylated glycoprotein with secretory capacity, was significantly upregulated in HCC and had an excellent correlation with patient survival and malignancy features. Through database mining and experimental validation, we found that TUFT1 was associated with fatty acid metabolism and promoted lipid accumulation in HCC cells. Further, we found that TUFT1 can interact with CREB1, a transcription factor for hepatic lipid metabolism, and regulate its activity and the transcriptions of key enzymes for lipogenesis. TUFT1 promoted HCC cell proliferation significantly, which was partially reversed by treatment of an inhibitor of CREB1, KG-501. Moreover, TUFT1 promoted the capacity of HCC cell invasion in vitro, which was likely mediated by its association with zyxin, a zinc-binding phosphoprotein responsible for the formation of fully mature focal adhesions on extracellular matrix. We found that TUFT1 can interact with ZYX and inhibit its expression and recruitments to focal complexes in HCC cells. Collectively, our study uncovered new regulatory mechanisms of TUFT1-mediated lipogenesis, cell proliferation, and invasion.
Collapse
|
13
|
Zhou H, Wang M, Zhang Y, Su Q, Xie Z, Chen X, Yan R, Li P, Li T, Qin X, Yang H, Wu C, You F, Li S, Liu Y. Functions and clinical significance of mechanical tumor microenvironment: cancer cell sensing, mechanobiology and metastasis. Cancer Commun (Lond) 2022; 42:374-400. [PMID: 35470988 PMCID: PMC9118059 DOI: 10.1002/cac2.12294] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/16/2022] [Accepted: 04/19/2022] [Indexed: 12/12/2022] Open
Abstract
Dynamic and heterogeneous interaction between tumor cells and the surrounding microenvironment fuels the occurrence, progression, invasion, and metastasis of solid tumors. In this process, the tumor microenvironment (TME) fractures cellular and matrix architecture normality through biochemical and mechanical means, abetting tumorigenesis and treatment resistance. Tumor cells sense and respond to the strength, direction, and duration of mechanical cues in the TME by various mechanotransduction pathways. However, far less understood is the comprehensive perspective of the functions and mechanisms of mechanotransduction. Due to the great therapeutic difficulties brought by the mechanical changes in the TME, emerging studies have focused on targeting the adverse mechanical factors in the TME to attenuate disease rather than conventionally targeting tumor cells themselves, which has been proven to be a potential therapeutic approach. In this review, we discussed the origins and roles of mechanical factors in the TME, cell sensing, mechano‐biological coupling and signal transduction, in vitro construction of the tumor mechanical microenvironment, applications and clinical significance in the TME.
Collapse
Affiliation(s)
- Hanying Zhou
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610054, P. R. China
| | - Meng Wang
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610054, P. R. China
| | - Yixi Zhang
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610054, P. R. China
| | - Qingqing Su
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610054, P. R. China
| | - Zhengxin Xie
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610054, P. R. China
| | - Xiangyan Chen
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610054, P. R. China
| | - Ran Yan
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610054, P. R. China.,Traditional Chinese Medicine Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610072, P. R. China
| | - Ping Li
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610054, P. R. China
| | - Tingting Li
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610054, P. R. China
| | - Xiang Qin
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610054, P. R. China
| | - Hong Yang
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610054, P. R. China
| | - Chunhui Wu
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610054, P. R. China
| | - Fengming You
- Traditional Chinese Medicine Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610072, P. R. China
| | - Shun Li
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610054, P. R. China
| | - Yiyao Liu
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610054, P. R. China.,Traditional Chinese Medicine Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610072, P. R. China
| |
Collapse
|
14
|
Oliveira-Rizzo C, Ottati MC, Fort RS, Chavez S, Trinidad JM, DiPaolo A, Garat B, Sotelo-Silveira JR, Duhagon MA. Hsa-miR-183-5p Modulates Cell Adhesion by Repression of ITGB1 Expression in Prostate Cancer. Noncoding RNA 2022; 8:ncrna8010011. [PMID: 35202085 PMCID: PMC8875343 DOI: 10.3390/ncrna8010011] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/05/2022] [Accepted: 01/08/2022] [Indexed: 12/24/2022] Open
Abstract
Prostate cancer is a major health problem worldwide. MiR-183 is an oncomiR and a candidate biomarker in prostate cancer, affecting various pathways responsible for disease initiation and progression. We sought to discover the most relevant processes controlled by miR-183 through an unbiased transcriptomic approach using prostate cell lines and patient tissues to identify miR-183 responsive genes and pathways. Gain of function experiments, reporter gene assays, and transcript and protein measurements were conducted to validate predicted functional effects and protein mediators. A total of 135 candidate miR-183 target genes overrepresenting cell adhesion terms were inferred from the integrated transcriptomic analysis. Cell attachment, spreading assays and focal adhesion quantification of miR-183-overexpressing cells confirmed the predicted reduction in cell adhesion. ITGB1 was validated as a major target of repression by miR-183 as well as a mediator of cell adhesion in response to miR-183. The reporter gene assay and PAR-CLIP read mapping suggest that ITGB1 may be a direct target of miR-183. The negative correlation between miR-183 and ITGB1 expression in prostate cancer cohorts supports their interaction in the clinical set. Overall, cell adhesion was uncovered as a major pathway controlled by miR-183 in prostate cancer, and ITGB1 was identified as a relevant mediator of this effect.
Collapse
Affiliation(s)
- Carolina Oliveira-Rizzo
- Laboratorio de Interacciones Moleculares, Facultad de Ciencias, Universidad de la República, Iguá 4225, Montevideo 11400, Uruguay; (C.O.-R.); (M.C.O.); (R.S.F.); (S.C.); (J.M.T.); (B.G.)
- Departamento de Genética, Facultad de Medicina, Universidad de la República, Av. Gral. Flores 2125, Montevideo 11800, Uruguay
| | - María Carolina Ottati
- Laboratorio de Interacciones Moleculares, Facultad de Ciencias, Universidad de la República, Iguá 4225, Montevideo 11400, Uruguay; (C.O.-R.); (M.C.O.); (R.S.F.); (S.C.); (J.M.T.); (B.G.)
- Departamento de Genética, Facultad de Medicina, Universidad de la República, Av. Gral. Flores 2125, Montevideo 11800, Uruguay
| | - Rafael Sebastián Fort
- Laboratorio de Interacciones Moleculares, Facultad de Ciencias, Universidad de la República, Iguá 4225, Montevideo 11400, Uruguay; (C.O.-R.); (M.C.O.); (R.S.F.); (S.C.); (J.M.T.); (B.G.)
- Departamento de Genética, Facultad de Medicina, Universidad de la República, Av. Gral. Flores 2125, Montevideo 11800, Uruguay
- Departamento de Genómica, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo 11600, Uruguay; (A.D.); (J.R.S.-S.)
| | - Santiago Chavez
- Laboratorio de Interacciones Moleculares, Facultad de Ciencias, Universidad de la República, Iguá 4225, Montevideo 11400, Uruguay; (C.O.-R.); (M.C.O.); (R.S.F.); (S.C.); (J.M.T.); (B.G.)
- Departamento de Genética, Facultad de Medicina, Universidad de la República, Av. Gral. Flores 2125, Montevideo 11800, Uruguay
- Departamento de Genómica, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo 11600, Uruguay; (A.D.); (J.R.S.-S.)
| | - Juan Manuel Trinidad
- Laboratorio de Interacciones Moleculares, Facultad de Ciencias, Universidad de la República, Iguá 4225, Montevideo 11400, Uruguay; (C.O.-R.); (M.C.O.); (R.S.F.); (S.C.); (J.M.T.); (B.G.)
- Departamento de Genética, Facultad de Medicina, Universidad de la República, Av. Gral. Flores 2125, Montevideo 11800, Uruguay
- Departamento de Genómica, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo 11600, Uruguay; (A.D.); (J.R.S.-S.)
| | - Andrés DiPaolo
- Departamento de Genómica, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo 11600, Uruguay; (A.D.); (J.R.S.-S.)
| | - Beatriz Garat
- Laboratorio de Interacciones Moleculares, Facultad de Ciencias, Universidad de la República, Iguá 4225, Montevideo 11400, Uruguay; (C.O.-R.); (M.C.O.); (R.S.F.); (S.C.); (J.M.T.); (B.G.)
| | - José Roberto Sotelo-Silveira
- Departamento de Genómica, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo 11600, Uruguay; (A.D.); (J.R.S.-S.)
- Departamento de Biología Celular, Facultad de Ciencias, Universidad de la República, Iguá 4225, Montevideo 11400, Uruguay
| | - María Ana Duhagon
- Laboratorio de Interacciones Moleculares, Facultad de Ciencias, Universidad de la República, Iguá 4225, Montevideo 11400, Uruguay; (C.O.-R.); (M.C.O.); (R.S.F.); (S.C.); (J.M.T.); (B.G.)
- Departamento de Genética, Facultad de Medicina, Universidad de la República, Av. Gral. Flores 2125, Montevideo 11800, Uruguay
- Correspondence: or ; Tel.: +598-2-525-8618 (ext. 7237) or +598-2-924-3414 (ext. 3468)
| |
Collapse
|
15
|
Atakhani A, Bogdziewiez L, Verger S. Characterising the mechanics of cell-cell adhesion in plants. QUANTITATIVE PLANT BIOLOGY 2022; 3:e2. [PMID: 37077973 PMCID: PMC10095952 DOI: 10.1017/qpb.2021.16] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 12/07/2021] [Accepted: 12/09/2021] [Indexed: 05/03/2023]
Abstract
Cell-cell adhesion is a fundamental feature of multicellular organisms. To ensure multicellular integrity, adhesion needs to be tightly controlled and maintained. In plants, cell-cell adhesion remains poorly understood. Here, we argue that to be able to understand how cell-cell adhesion works in plants, we need to understand and quantitatively measure the mechanics behind it. We first introduce cell-cell adhesion in the context of multicellularity, briefly explain the notions of adhesion strength, work and energy and present the current knowledge concerning the mechanisms of cell-cell adhesion in plants. Because still relatively little is known in plants, we then turn to animals, but also algae, bacteria, yeast and fungi, and examine how adhesion works and how it can be quantitatively measured in these systems. From this, we explore how the mechanics of cell adhesion could be quantitatively characterised in plants, opening future perspectives for understanding plant multicellularity.
Collapse
Affiliation(s)
- Asal Atakhani
- Department of Forest Genetics and Plant Physiology, Umeå Plant Science Centre, Swedish University of Agricultural Sciences, Umeå, Sweden
| | - Léa Bogdziewiez
- Department of Forest Genetics and Plant Physiology, Umeå Plant Science Centre, Swedish University of Agricultural Sciences, Umeå, Sweden
| | - Stéphane Verger
- Department of Forest Genetics and Plant Physiology, Umeå Plant Science Centre, Swedish University of Agricultural Sciences, Umeå, Sweden
- Author for correspondence: S. Verger, E-mail:
| |
Collapse
|
16
|
Song Y, Long J, Dunkers JP, Woodcock JW, Lin H, Fox DM, Liao X, Lv Y, Yang L, Chiang MYM. Micromechanical Compatibility between Cells and Scaffolds Directs the Phenotypic Transition of Stem Cells. ACS APPLIED MATERIALS & INTERFACES 2021; 13:58152-58161. [PMID: 34808061 DOI: 10.1021/acsami.1c17504] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
This study experimentally substantiates that the micromechanical compatibility between cell and substrate is essential for cells to achieve energetically favorable mechanotransduction that directs phenotypic transitions. The argument for this compatibility is based on a thermodynamic model that suggests that the response of cells to their substrate mechanical environment is a consequence of the interchange between forms of energy governing the cell-substrate interaction. Experimental validation for the model has been carried out by investigating the osteogenic differentiation of dental follicle stem cells (DFSCs) seeded on electrospun fibrous scaffolds. Electrospinning of blends containing polycaprolactone (PCL) and silk fibroin (SF) with varying composition of cellulose nanocrystals (CNCs) resulted in three-dimensional (3D) fibrous scaffolds with bimodal distribution of fiber diameter, which provides both macroscopically stiff and microscopically compliant scaffolds for cells without affecting the surface chemical functionality of scaffolds. Atomic force microscopy (AFM) with a colloidal probe and single-cell force spectroscopy were used to characterize cell stiffness and scaffold stiffness on the cellular level, as well as cell-scaffold adhesive interaction (chemical functionality). This study has successfully varied scaffold mechanical properties without affecting their surface chemistry. In vitro tests indicate that the micromechanical compatibility between cells and scaffolds has been significantly correlated with mechanosensitive gene expression markers and osteogenic differentiation markers of DFSCs. The agreement between experimental observations and the thermodynamic model affirms that the cellular response to the mechanical environment, though biological in nature, follows the laws of the energy interchange to achieve its self-regulating behavior. More importantly, this study provides systematic evidence, through extensive and rigorous experimental studies, for the first time that rationalizes that micromechanical compatibility is indeed important to the efficacy of regenerative medicine.
Collapse
Affiliation(s)
- Yang Song
- Materials Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
- 111 Project Laboratory of Biomechanics and Tissue Repair, Bioengineering College, Chongqing University, Chongqing 400044, China
| | - Jiaoyue Long
- Materials Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
- Kangcell Biotechnology, Chongqing 400714, China
| | - Joy P Dunkers
- Materials Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
| | - Jeremiah W Woodcock
- Materials Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
| | - Hungchun Lin
- Materials Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
| | - Douglas M Fox
- Department of Chemistry, American University, Washington, District of Columbia 20016, United States
| | - Xiaoling Liao
- Chongqing Key Laboratory of Nano/Micro Composite Materials and Devices, Chongqing University of Science and Technology, Chongqing 400050, China
| | - Yonggang Lv
- 111 Project Laboratory of Biomechanics and Tissue Repair, Bioengineering College, Chongqing University, Chongqing 400044, China
| | - Li Yang
- 111 Project Laboratory of Biomechanics and Tissue Repair, Bioengineering College, Chongqing University, Chongqing 400044, China
| | - Martin Y M Chiang
- Materials Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
| |
Collapse
|
17
|
De Luca M, Mandala M, Rose G. Towards an understanding of the mechanoreciprocity process in adipocytes and its perturbation with aging. Mech Ageing Dev 2021; 197:111522. [PMID: 34147549 DOI: 10.1016/j.mad.2021.111522] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 05/29/2021] [Accepted: 06/15/2021] [Indexed: 12/25/2022]
Abstract
Adipose tissue (AT) is a complex organ, with multiple functions that are essential for maintaining metabolic health. A feature of AT is its capability to expand in response to physiological challenges, such as pregnancy and aging, and during chronic states of positive energy balance occurring throughout life. AT grows through adipogenesis and/or an increase in the size of existing adipocytes. One process that is required for healthy AT growth is the remodeling of the extracellular matrix (ECM), which is a necessary step to restore mechanical homeostasis and maintain tissue integrity and functionality. While the relationship between mechanobiology and adipogenesis is now well recognized, less is known about the role of adipocyte mechanosignaling pathways in AT growth. In this review article, we first summarize evidence linking ECM remodelling to AT expansion and how its perturbation is associated to a metabolically unhealthy phenotype. Subsequently, we highlight findings suggesting that molecules involved in the dynamic, bidirectional process (mechanoreciprocity) enabling adipocytes to sense changes in the mechanical properties of the ECM are interconnected to pathways regulating lipid metabolism. Finally, we discuss processes through which aging may influence the ability of adipocytes to appropriately respond to alterations in ECM composition.
Collapse
Affiliation(s)
- Maria De Luca
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Maurizio Mandala
- Department of Biology, Ecology and Earth Science, University of Calabria, Rende, 87036, Italy
| | - Giuseppina Rose
- Department of Biology, Ecology and Earth Science, University of Calabria, Rende, 87036, Italy
| |
Collapse
|
18
|
Chen S, Liu A, Wu C, Chen Y, Liu C, Zhang Y, Wu K, Wei D, Sun J, Zhou L, Fan H. Static-Dynamic Profited Viscoelastic Hydrogels for Motor-Clutch-Regulated Neurogenesis. ACS APPLIED MATERIALS & INTERFACES 2021; 13:24463-24476. [PMID: 34024102 DOI: 10.1021/acsami.1c03821] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Viscoelasticity, a time-scale mechanical feature of the native extracellular matrix (ECM), is reported to play crucial roles in plentiful cellular behaviors, whereas its effects on neuronal behavior and the underlying molecular mechanism still remain obscure. Challenges are faced in the biocompatible synthesis of neural ECM-mimicked scaffolds solely controlled with viscoelasticity and due to the lack of suitable models for neurons-viscoelastic matrix interaction. Herein, we report difunctional hyaluronan-collagen hydrogels prepared by a static-dynamic strategy. The hydrogels show aldehyde concentration-dependent viscoelasticity and similar initial elastic modulus, fibrillar morphology, swelling as well as degradability. Utilizing the resulting hydrogels, for the first time, we demonstrate matrix viscoelasticity-dependent neuronal responses, including neurite elongation and expression of neurogenic proteins. Then, a motor-clutch model modified with a tension dissipation component is developed to account for the molecular mechanism for viscoelasticity-sensitive neuronal responses. Moreover, we prove enhanced recovery of rat spinal cord injury by implanting cell-free viscoelastic grafts. As a pioneer finding on neurons-viscoelastic matrix interaction both in vitro and in vivo, this work provides intriguing insights not only into nerve repair but also into neuroscience and tissue engineering.
Collapse
Affiliation(s)
- Suping Chen
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064 Sichuan, China
| | - Amin Liu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064 Sichuan, China
| | - Chengheng Wu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064 Sichuan, China
| | - Yaxing Chen
- Department of Neurosurgery, West China Medical School, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China
| | - Chang Liu
- Department of Neurosurgery, West China Medical School, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China
| | - Yusheng Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064 Sichuan, China
| | - Kai Wu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064 Sichuan, China
| | - Dan Wei
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064 Sichuan, China
| | - Jing Sun
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064 Sichuan, China
| | - Liangxue Zhou
- Department of Neurosurgery, West China Medical School, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China
| | - Hongsong Fan
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064 Sichuan, China
| |
Collapse
|
19
|
Xie P, Yuan FQ, Huang MS, Zhang W, Zhou HH, Li X, Liu ZQ. DCBLD2 Affects the Development of Colorectal Cancer via EMT and Angiogenesis and Modulates 5-FU Drug Resistance. Front Cell Dev Biol 2021; 9:669285. [PMID: 34095137 PMCID: PMC8170045 DOI: 10.3389/fcell.2021.669285] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 04/27/2021] [Indexed: 01/05/2023] Open
Abstract
Background: DCBLD2 is highly expressed in various cancers, including colorectal cancer. DCBLD2 overexpression promotes tumor occurrence, development, and metastasis. However, DCBLD2 sensitivity to chemotherapy drugs and its mechanism on tumor development are unknown. Methods: DCBLD2 expression differences in cancer and normal tissues were obtained from GEO and TCGA databases. DCBLD2 influence on prognosis was also compared, and the database analysis results were verified via the analysis of clinical samples. GDSC database was used to analyze the effect of DCBLD2 expression difference on 5-FU drug sensitivity on tumor cells. CCK-8, clone formation, scratch, Transwell invasion and migration assays were used to assess DCBLD2 effects on the proliferation, metastasis, and 5-FU drug sensitivity on HCT116 and Caco-2 colorectal cancer cells. Angiogenesis and Matrigel plug assays were used to study the effect of DCBLD2 on angiogenesis. Q-RCR and Western Blot were used to analyze DCBLD2 impact on the EMT signaling pathway, and TAP-MS assay with Co-IP verification was used to identify the downstream target proteins binding to DCBLD2. Results: Both database and clinical sample validation results showed that the expression of DCBLD2 in colorectal cancer tissues was significantly higher than that in normal tissues, leading to poor prognosis of patients. GDSC database analysis showed that DCBLD2 overexpression caused tumor cell resistance to 5-FU. The results of in vitro and in vivo experiments showed that the inhibition of DCBLD2 reduced the proliferation, migration and invasion of colorectal cancer cells, inhibited the angiogenesis of endothelial cells, and enhanced the drug sensitivity to 5-FU. The results of q-RCR and Western Blot experiments showed that the inhibition of DCBLD2 can suppress the EMT signal. The results of TAP-MS assay showed that the proteins bound to DCBLD2 were enriched to the Focal adhesion pathway. The results of Co-IP assay show that DCBLD2 can combine with ITGB1, the key factor of Focal adhesion pathway. Conclusion: DCBLD2 may affect the development of colorectal cancer by regulating cell proliferation and motility, and modulate 5-FU resistance. Down-regulation of DCBLD2 can inhibit EMT signal and angiogenesis. DCBLD2 can combine with ITGB1, the key signal factor of the Focal adhesion pathway.
Collapse
Affiliation(s)
- Pan Xie
- Hunan Key Laboratory of Pharmacogenetics, Department of Clinical Pharmacology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Fu-Qiang Yuan
- Hunan Key Laboratory of Pharmacogenetics, Department of Clinical Pharmacology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Ma-Sha Huang
- Hunan Key Laboratory of Pharmacogenetics, Department of Clinical Pharmacology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Wei Zhang
- Hunan Key Laboratory of Pharmacogenetics, Department of Clinical Pharmacology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Hong-Hao Zhou
- Hunan Key Laboratory of Pharmacogenetics, Department of Clinical Pharmacology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Xi Li
- Hunan Key Laboratory of Pharmacogenetics, Department of Clinical Pharmacology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Zhao-Qian Liu
- Hunan Key Laboratory of Pharmacogenetics, Department of Clinical Pharmacology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Central South University, Changsha, China
| |
Collapse
|
20
|
Hernández-Cáceres MP, Munoz L, Pradenas JM, Pena F, Lagos P, Aceiton P, Owen GI, Morselli E, Criollo A, Ravasio A, Bertocchi C. Mechanobiology of Autophagy: The Unexplored Side of Cancer. Front Oncol 2021; 11:632956. [PMID: 33718218 PMCID: PMC7952994 DOI: 10.3389/fonc.2021.632956] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 02/01/2021] [Indexed: 12/12/2022] Open
Abstract
Proper execution of cellular function, maintenance of cellular homeostasis and cell survival depend on functional integration of cellular processes and correct orchestration of cellular responses to stresses. Cancer transformation is a common negative consequence of mismanagement of coordinated response by the cell. In this scenario, by maintaining the balance among synthesis, degradation, and recycling of cytosolic components including proteins, lipids, and organelles the process of autophagy plays a central role. Several environmental stresses activate autophagy, among those hypoxia, DNA damage, inflammation, and metabolic challenges such as starvation. In addition to these chemical challenges, there is a requirement for cells to cope with mechanical stresses stemming from their microenvironment. Cells accomplish this task by activating an intrinsic mechanical response mediated by cytoskeleton active processes and through mechanosensitive protein complexes which interface the cells with their mechano-environment. Despite autophagy and cell mechanics being known to play crucial transforming roles during oncogenesis and malignant progression their interplay is largely overlooked. In this review, we highlight the role of physical forces in autophagy regulation and their potential implications in both physiological as well as pathological conditions. By taking a mechanical perspective, we wish to stimulate novel questions to further the investigation of the mechanical requirements of autophagy and appreciate the extent to which mechanical signals affect this process.
Collapse
Affiliation(s)
- Maria Paz Hernández-Cáceres
- Laboratory of Autophagy and Metabolism, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica De Chile, Santiago, Chile
| | - Leslie Munoz
- Laboratory for Mechanobiology of Transforming Systems, Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
- Laboratory for Molecular Mechanics of Cell Adhesion, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica De Chile, Santiago, Chile
| | - Javiera M. Pradenas
- Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile
- Laboratory of Investigation in Oncology, Faculty of Biological Sciences Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Francisco Pena
- Laboratory for Mechanobiology of Transforming Systems, Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
- Laboratory for Molecular Mechanics of Cell Adhesion, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica De Chile, Santiago, Chile
| | - Pablo Lagos
- Laboratory of Autophagy and Metabolism, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica De Chile, Santiago, Chile
| | - Pablo Aceiton
- Laboratory for Mechanobiology of Transforming Systems, Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
- Laboratory for Molecular Mechanics of Cell Adhesion, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica De Chile, Santiago, Chile
| | - Gareth I. Owen
- Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile
- Laboratory of Investigation in Oncology, Faculty of Biological Sciences Pontificia Universidad Católica de Chile, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Eugenia Morselli
- Laboratory of Autophagy and Metabolism, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica De Chile, Santiago, Chile
- Autophagy Research Center, Santiago de Chile, Chile
| | - Alfredo Criollo
- Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile
- Autophagy Research Center, Santiago de Chile, Chile
- Facultad De Odontología, Instituto De Investigación En Ciencias Odontológicas (ICOD), Universidad De Chile, Santiago, Chile
| | - Andrea Ravasio
- Laboratory for Mechanobiology of Transforming Systems, Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cristina Bertocchi
- Laboratory for Molecular Mechanics of Cell Adhesion, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica De Chile, Santiago, Chile
| |
Collapse
|
21
|
Wang J, Sui J, Mao C, Li X, Chen X, Liang C, Wang X, Wang SH, Jia C. Identification of Key Pathways and Genes Related to the Development of Hair Follicle Cycle in Cashmere Goats. Genes (Basel) 2021; 12:genes12020180. [PMID: 33513983 PMCID: PMC7911279 DOI: 10.3390/genes12020180] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 01/15/2021] [Accepted: 01/22/2021] [Indexed: 12/13/2022] Open
Abstract
The development of hair follicle in cashmere goats shows significant periodic change, as with mice and humans. However, for cashmere goat with double-coat, the periodic change may be due to other regulatory molecules and signal pathways. To understand the mechanism of periodic development of hair follicle, we performed a weighted gene coexpression network analysis (WGCNA) to mine key genes and establish an interaction network by utilizing the NCBI public dataset. Ten coexpression modules, including 7689 protein-coding genes, were constructed by WGCNA, six of which are considered to be significantly related to the development of the hair follicle cycle. A functional enrichment analysis for each model showed that they are closely related to ECM- receptor interaction, focal adhesion, PI3K-Akt signaling pathway, estrogen signaling pathway, and so on. Combined with the analysis of differential expressed genes, 12 hub genes from coexpression modules were selected as candidate markers, i.e., COL1A1, C1QTNF6, COL1A2, AQP3, KRTAP3-1, KRTAP11-1, FA2H, NDUFS5, DERL2, MRPL14, ANTKMT and XAB2, which might be applied to improve cashmere production.
Collapse
|
22
|
Juhl OJ, Merife AB, Zhang Y, Lemmon CA, Donahue HJ. Hydroxyapatite Particle Density Regulates Osteoblastic Differentiation Through β-Catenin Translocation. Front Bioeng Biotechnol 2021; 8:591084. [PMID: 33490047 PMCID: PMC7820766 DOI: 10.3389/fbioe.2020.591084] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 12/08/2020] [Indexed: 01/09/2023] Open
Abstract
Substrate surface characteristics such as roughness, wettability and particle density are well-known contributors of a substrate's overall osteogenic potential. These characteristics are known to regulate cell mechanics as well as induce changes in cell stiffness, cell adhesions, and cytoskeletal structure. Pro-osteogenic particles, such as hydroxyapatite, are often incorporated into a substrate to enhance the substrates osteogenic potential. However, it is unknown which substrate characteristic is the key regulator of osteogenesis. This is partly due to the lack of understanding of how these substrate surface characteristics are transduced by cells. In this study substrates composed of polycaprolactone (PCL) and carbonated hydroxyapatite particles (HAp) were synthesized. HAp concentration was varied, and a range of surface characteristics created. The effect of each substrate characteristic on osteoblastic differentiation was then examined. We found that, of the characteristics examined, only HAp density, and indeed a specific density (85 particles/cm2), significantly increased osteoblastic differentiation. Further, an increase in focal adhesion maturation and turnover was observed in cells cultured on this substrate. Moreover, β-catenin translocation from the membrane bound cell fraction to the nucleus was more rapid in cells on the 85 particle/cm2 substrate compared to cells on tissue culture polystyrene. Together, these data suggest that particle density is one pivotal factor in determining a substrates overall osteogenic potential. Additionally, the observed increase in osteoblastic differentiation is a at least partly the result of β-catenin translocation and transcriptional activity suggesting a β-catenin mediated mechanism by which substrate surface characteristics are transduced.
Collapse
Affiliation(s)
- Otto J Juhl
- Department of Biomedical Engineering and Institute for Engineering and Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Anna-Blessing Merife
- Department of Biomedical Engineering and Institute for Engineering and Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Yue Zhang
- Department of Biomedical Engineering and Institute for Engineering and Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Christopher A Lemmon
- Department of Biomedical Engineering and Institute for Engineering and Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Henry J Donahue
- Department of Biomedical Engineering and Institute for Engineering and Medicine, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
23
|
Natural and Synthetic Biomaterials for Engineering Multicellular Tumor Spheroids. Polymers (Basel) 2020; 12:polym12112506. [PMID: 33126468 PMCID: PMC7692845 DOI: 10.3390/polym12112506] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/23/2020] [Accepted: 10/26/2020] [Indexed: 02/06/2023] Open
Abstract
The lack of in vitro models that represent the native tumor microenvironment is a significant challenge for cancer research. Two-dimensional (2D) monolayer culture has long been the standard for in vitro cell-based studies. However, differences between 2D culture and the in vivo environment have led to poor translation of cancer research from in vitro to in vivo models, slowing the progress of the field. Recent advances in three-dimensional (3D) culture have improved the ability of in vitro culture to replicate in vivo conditions. Although 3D cultures still cannot achieve the complexity of the in vivo environment, they can still better replicate the cell-cell and cell-matrix interactions of solid tumors. Multicellular tumor spheroids (MCTS) are three-dimensional (3D) clusters of cells with tumor-like features such as oxygen gradients and drug resistance, and represent an important translational tool for cancer research. Accordingly, natural and synthetic polymers, including collagen, hyaluronic acid, Matrigel®, polyethylene glycol (PEG), alginate and chitosan, have been used to form and study MCTS for improved clinical translatability. This review evaluates the current state of biomaterial-based MCTS formation, including advantages and disadvantages of the different biomaterials and their recent applications to the field of cancer research, with a focus on the past five years.
Collapse
|
24
|
Zeng Y, Cao Y, Liu L, Zhao J, Zhang T, Xiao L, Jia M, Tian Q, Yu H, Chen S, Cai Y. SEPT9_i1 regulates human breast cancer cell motility through cytoskeletal and RhoA/FAK signaling pathway regulation. Cell Death Dis 2019; 10:720. [PMID: 31558699 PMCID: PMC6763430 DOI: 10.1038/s41419-019-1947-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 08/09/2019] [Accepted: 08/26/2019] [Indexed: 02/08/2023]
Abstract
Increasing cell mobility is the basis of tumor invasion and metastasis, and is therefore a therapeutic target for preventing the spread of many types of cancer. Septins are a family of cytoskeletal proteins with GTPase activity, and play a role in many important cellular functions, including cell migration. SEPT9 isoform 1 protein (SEPT9_i1) has been associated with breast tumor development and the enhancement of cell migration; however, the exact mechanism of how SEPT9_i1 might affect breast cancer progression remains to be elucidated. Here, we report that the expression of SEPT9_i1 positively correlated with paxillin, and both were significantly upregulated in invasive breast cancer tissues of patients with lymph node metastases. Lentivirus-mediated shRNA knockdown of SEPT9 in MCF-7 cells diminished tumor cell migration, focal adhesion (FA) maturation and the expression of β-actin, β-tubulin, Cdc42, RhoA, and Rac, whereas overexpression of SEPT9_i1 in SEPT9-knockdown MCF-7 cells promoted cell migration, FA maturation and relevant protein expression. Furthermore, overexpression of SEPT9_i1 in MCF-7 cells markedly increased FAK/Src/paxillin signaling, at least in part through RhoA/ROCK1 upstream activation. Transcriptome profiling suggested that SEPT9_i1 may directly affect “Focal adhesion” and “Regulation of actin cytoskeleton” signaling mechanisms. Finally, overexpression of SEPT9_i1 markedly enhanced lung metastases in vivo 6 weeks after tumor inoculation. These findings suggest that a mechanism of Septin-9-induced aberrant cancer cell migration is through cytoskeletal regulation and FA modulation, and encourages the use of SEPT9 as novel therapeutic target in the prevention of tumor metastasis.
Collapse
Affiliation(s)
- Yongqiu Zeng
- Key Laboratory of Obstetric, Gynecologic, and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu, Sichuan, China. .,Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China.
| | - Yang Cao
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Lan Liu
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Jiao Zhao
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Ting Zhang
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Lifan Xiao
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Man Jia
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Qiang Tian
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Hong Yu
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Shaokun Chen
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Yansen Cai
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
25
|
Abstract
The pulmonary endothelial cell forms a critical semi-permeable barrier between the vascular and interstitial space. As part of the blood-gas barrier in the lung, the endothelium plays a key role in normal physiologic function and pathologic disease. Changes in endothelial cell shape, defined by its plasma membrane, determine barrier integrity. A number of key cytoskeletal regulatory and effector proteins including non-muscle myosin light chain kinase, cortactin, and Arp 2/3 mediate actin rearrangements to form cortical and membrane associated structures in response to barrier enhancing stimuli. These actin formations support and interact with junctional complexes and exert forces to protrude the lipid membrane to and close gaps between individual cells. The current knowledge of these cytoskeletal processes and regulatory proteins are the subject of this review. In addition, we explore novel advancements in cellular imaging that are poised to shed light on the complex nature of pulmonary endothelial permeability.
Collapse
|
26
|
Abstract
Reproductive biologists are well-versed in many types of biochemical signaling, and indeed, there are almost innumerable examples in reproduction, including steroid and peptide hormone signaling, receptor-ligand and secondary messenger-mediated signaling, signaling regulated by membrane channels, and many others. Among reproductive scientists, a perhaps lesser-known but comparably important mode of signaling is mechanotransduction: the concept that cells can sense and respond to externally applied or internally generated mechanical forces. Given the cell shape changes and tissue morphogenesis events that are components of many phenomena in reproductive function, it should be no surprise that mechanotransduction has major impacts in reproductive health and pathophysiology. The conference on "Mechanotransduction in the Reproductive Tract" was a valuable launch pad to bring this hot issue in development, cell biology, biophysics, and tissue regeneration to the realm of reproductive biology. The goal of the meeting was to stimulate interest and increased mechanotransduction research in the reproductive field by presenting a broad spectrum of responses impacted by this process. The meeting highlighted the importance of convening expert investigators, students, fellows, and young investigators from a number of research areas resulting in cross-fertilization of ideas and suggested new avenues for study. The conference included talks on tissue engineering, stem cells, and several areas of reproductive biology, from uterus and cervix to the gametes. Specific reproductive health-relevant areas, including uterine fibroids, gestation and parturition, and breast tissue morphogenesis, received particular attention.
Collapse
Affiliation(s)
- Janice P Evans
- a Department of Biochemistry and Molecular Biology , Bloomberg School of Public Health, Johns Hopkins University , Baltimore , MD , USA
| | - Phyllis C Leppert
- b Department of Obstetrics and Gynecology , Duke University School of Medicine , Durham , NC , USA.,c The Campion Fund , Durham , NC , USA
| |
Collapse
|
27
|
Steeves AJ, Atwal A, Schock SC, Variola F. Evaluation of the direct effects of poly(dopamine) on the in vitro response of human osteoblastic cells. J Mater Chem B 2016; 4:3145-3156. [DOI: 10.1039/c5tb02510a] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Functional poly(dopamine) coatings promise to become an efficient strategy to endow biomaterials with enhanced bioactive properties.
Collapse
Affiliation(s)
- Alexander J. Steeves
- Faculty of Engineering
- Department of Mechanical Engineering
- University of Ottawa
- Canada
| | - Aman Atwal
- Faculty of Science
- Department of Biopharmaceutical Sciences
- University of Ottawa
- Canada
| | - Sarah C. Schock
- The Children's Hospital of Eastern Ontario (CHEO) Research Institute
- Canada
- Faculty of Medicine
- Department of Cellular and Molecular Medicine
- University of Ottawa
| | - Fabio Variola
- Faculty of Engineering
- Department of Mechanical Engineering
- University of Ottawa
- Canada
- Faculty of Medicine
| |
Collapse
|
28
|
Focal adhesion kinase-dependent focal adhesion recruitment of SH2 domains directs SRC into focal adhesions to regulate cell adhesion and migration. Sci Rep 2015; 5:18476. [PMID: 26681405 PMCID: PMC4683442 DOI: 10.1038/srep18476] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 11/19/2015] [Indexed: 01/09/2023] Open
Abstract
Directed cell migration requires dynamical control of the protein complex within focal adhesions (FAs) and this control is regulated by signaling events involving tyrosine phosphorylation. We screened the SH2 domains present in tyrosine-specific kinases and phosphatases found within FAs, including SRC, SHP1 and SHP2, and examined whether these enzymes transiently target FAs via their SH2 domains. We found that the SRC_SH2 domain and the SHP2_N-SH2 domain are associated with FAs, but only the SRC_SH2 domain is able to be regulated by focal adhesion kinase (FAK). The FAK-dependent association of the SRC_SH2 domain is necessary and sufficient for SRC FA targeting. When the targeting of SRC into FAs is inhibited, there is significant suppression of SRC-mediated phosphorylation of paxillin and FAK; this results in an inhibition of FA formation and maturation and a reduction in cell migration. This study reveals an association between FAs and the SRC_SH2 domain as well as between FAs and the SHP2_N-SH2 domains. This supports the hypothesis that the FAK-regulated SRC_SH2 domain plays an important role in directing SRC into FAs and that this SRC-mediated FA signaling drives cell migration.
Collapse
|
29
|
Pasipoularides A. Mechanotransduction Mechanisms for Intraventricular Diastolic Vortex Forces and Myocardial Deformations: Part 2. J Cardiovasc Transl Res 2015; 8:293-318. [PMID: 25971844 PMCID: PMC4519381 DOI: 10.1007/s12265-015-9630-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 04/27/2015] [Indexed: 01/10/2023]
Abstract
Epigenetic mechanisms are fundamental in cardiac adaptations, remodeling, reverse remodeling, and disease. A primary goal of translational cardiovascular research is recognizing whether disease-related changes in phenotype can be averted by eliminating or reducing the effects of environmental epigenetic risks. There may be significant medical benefits in using gene-by-environment interaction knowledge to prevent or reverse organ abnormalities and disease. This survey proposes that "environmental" forces associated with diastolic RV/LV rotatory flows exert important, albeit still unappreciated, epigenetic actions influencing functional and morphological cardiac adaptations. Mechanisms analogous to Murray's law of hydrodynamic shear-induced endothelial cell modulation of vascular geometry are likely to link diastolic vortex-associated shear, torque and "squeeze" forces to RV/LV adaptations. The time has come to explore a new paradigm in which such forces play a fundamental epigenetic role, and to work out how heart cells react to them. Findings from various imaging modalities, computational fluid dynamics, molecular cell biology and cytomechanics are considered. The following are examined, among others: structural dynamics of myocardial cells (endocardium, cardiomyocytes, and fibroblasts), cytoskeleton, nucleoskeleton, and extracellular matrix; mechanotransduction and signaling; and mechanical epigenetic influences on genetic expression. To help integrate and focus relevant pluridisciplinary research, rotatory RV/LV filling flow is placed within a working context that has a cytomechanics perspective. This new frontier in cardiac research should uncover versatile mechanistic insights linking filling vortex patterns and attendant forces to variable expressions of gene regulation in RV/LV myocardium. In due course, it should reveal intrinsic homeostatic arrangements that support ventricular myocardial function and adaptability.
Collapse
Affiliation(s)
- Ares Pasipoularides
- Department of Surgery, Duke University School of Medicine, Durham, NC, 27710, USA,
| |
Collapse
|