1
|
Huang Z, Gustave W, Bai S, Li Y, Li B, Elçin E, Jiang B, Jia Z, Zhang X, Shaheen SM, He F. Challenges and opportunities in commercializing whole-cell bioreporters in environmental application. ENVIRONMENTAL RESEARCH 2024; 262:119801. [PMID: 39147190 DOI: 10.1016/j.envres.2024.119801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/08/2024] [Accepted: 08/13/2024] [Indexed: 08/17/2024]
Abstract
Since the initial introduction of whole-cell bioreporters (WCBs) nearly 30 years ago, their high sensitivity, selectivity, and suitability for on-site detection have rendered them highly promising for environmental monitoring, medical diagnosis, food safety, biomanufacturing, and other fields. Especially in the environmental field, the technology provides a fast and efficient way to assess the bioavailability of pollutants in the environment. Despite these advantages, the technology has not been commercialized. This lack of commercialization is confusing, given the broad application prospects of WCBs. Over the years, numerous research papers have focused primarily on enhancing the sensitivity and selectivity of WCBs, with little attention paid to their wider commercial applications. So far, there is no a critical review has been published yet on this topic. Therefore, in this article we critically reviewed the research progress of WCBs over the past three decades, assessing the performance and limitations of current systems to understand the barriers to commercial deployment. By identifying these obstacles, this article provided researchers and industry stakeholders with deeper insights into the challenges hindering market entry and inspire further research toward overcoming these barriers, thereby facilitating the commercialization of WCBs as a promising technology for environmental monitoring.
Collapse
Affiliation(s)
- Zefeng Huang
- Institute of Environmental Processes and Pollution Control, School of Environment and Ecology, Jiangnan University, Wuxi, 214122, China
| | - Williamson Gustave
- School of Chemistry, Environmental & Life Sciences, University of the Bahamas, Nassau, 4912, Bahamas
| | - Shanshan Bai
- Institute of Environmental Processes and Pollution Control, School of Environment and Ecology, Jiangnan University, Wuxi, 214122, China
| | - Yongshuo Li
- Institute of Environmental Processes and Pollution Control, School of Environment and Ecology, Jiangnan University, Wuxi, 214122, China
| | - Boling Li
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou, Jiangsu, 215123, China; Meadows Center for Water and the Environment, Texas State University, San Marcos, TX, 78666, USA
| | - Evrim Elçin
- Department of Agricultural Biotechnology, Division of Enzyme and Microbial Biotechnology, Faculty of Agriculture, Aydın Adnan Menderes University, Aydın, 09970, Turkey
| | - Bo Jiang
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Zhemin Jia
- Institute of Environmental Processes and Pollution Control, School of Environment and Ecology, Jiangnan University, Wuxi, 214122, China
| | - Xiaokai Zhang
- Institute of Environmental Processes and Pollution Control, School of Environment and Ecology, Jiangnan University, Wuxi, 214122, China.
| | - Sabry M Shaheen
- University of Wuppertal, School of Architecture and Civil Engineering, Institute of Foundation Engineering, Water- and Waste-Management, Laboratory of Soil- and Groundwater-Management, Pauluskirchstraße 7, 42285 Wuppertal, Germany; King Abdulaziz University, Faculty of Environmental Sciences, Department of Agriculture, 21589 Jeddah, Saudi Arabia; University of Kafrelsheikh, Faculty of Agriculture, Department of Soil and Water Sciences, 33516, Kafr El-Sheikh, Egypt
| | - Feng He
- Institute of Environmental Processes and Pollution Control, School of Environment and Ecology, Jiangnan University, Wuxi, 214122, China
| |
Collapse
|
2
|
Celik B, Leal AF, Tomatsu S. Potential Targeting Mechanisms for Bone-Directed Therapies. Int J Mol Sci 2024; 25:8339. [PMID: 39125906 PMCID: PMC11312506 DOI: 10.3390/ijms25158339] [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: 05/21/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
Bone development is characterized by complex regulation mechanisms, including signal transduction and transcription factor-related pathways, glycobiological processes, cellular interactions, transportation mechanisms, and, importantly, chemical formation resulting from hydroxyapatite. Any abnormal regulation in the bone development processes causes skeletal system-related problems. To some extent, the avascularity of cartilage and bone makes drug delivery more challenging than that of soft tissues. Recent studies have implemented many novel bone-targeting approaches to overcome drawbacks. However, none of these strategies fully corrects skeletal dysfunction, particularly in growth plate-related ones. Although direct recombinant enzymes (e.g., Vimizim for Morquio, Cerezyme for Gaucher, Elaprase for Hunter, Mepsevii for Sly diseases) or hormone infusions (estrogen for osteoporosis and osteoarthritis), traditional gene delivery (e.g., direct infusion of viral or non-viral vectors with no modifications on capsid, envelope, or nanoparticles), and cell therapy strategies (healthy bone marrow or hematopoietic stem cell transplantation) partially improve bone lesions, novel delivery methods must be addressed regarding target specificity, less immunogenicity, and duration in circulation. In addition to improvements in bone delivery, potential regulation of bone development mechanisms involving receptor-regulated pathways has also been utilized. Targeted drug delivery using organic and inorganic compounds is a promising approach in mostly preclinical settings and future clinical translation. This review comprehensively summarizes the current bone-targeting strategies based on bone structure and remodeling concepts while emphasizing potential approaches for future bone-targeting systems.
Collapse
Affiliation(s)
- Betul Celik
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA;
- Nemours Children’s Health, 1600 Rockland Rd., Wilmington, DE 19803, USA;
| | - Andrés Felipe Leal
- Nemours Children’s Health, 1600 Rockland Rd., Wilmington, DE 19803, USA;
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá 110231, Colombia
| | - Shunji Tomatsu
- Nemours Children’s Health, 1600 Rockland Rd., Wilmington, DE 19803, USA;
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu 501-1193, Japan
- Department of Pediatrics, Thomas Jefferson University, Philadelphia, PA 19144, USA
| |
Collapse
|
3
|
Takács R, Kovács P, Ebeid RA, Almássy J, Fodor J, Ducza L, Barrett-Jolley R, Lewis R, Matta C. Ca2+-Activated K+ Channels in Progenitor Cells of Musculoskeletal Tissues: A Narrative Review. Int J Mol Sci 2023; 24:ijms24076796. [PMID: 37047767 PMCID: PMC10095002 DOI: 10.3390/ijms24076796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/01/2023] [Accepted: 04/04/2023] [Indexed: 04/08/2023] Open
Abstract
Musculoskeletal disorders represent one of the main causes of disability worldwide, and their prevalence is predicted to increase in the coming decades. Stem cell therapy may be a promising option for the treatment of some of the musculoskeletal diseases. Although significant progress has been made in musculoskeletal stem cell research, osteoarthritis, the most-common musculoskeletal disorder, still lacks curative treatment. To fine-tune stem-cell-based therapy, it is necessary to focus on the underlying biological mechanisms. Ion channels and the bioelectric signals they generate control the proliferation, differentiation, and migration of musculoskeletal progenitor cells. Calcium- and voltage-activated potassium (KCa) channels are key players in cell physiology in cells of the musculoskeletal system. This review article focused on the big conductance (BK) KCa channels. The regulatory function of BK channels requires interactions with diverse sets of proteins that have different functions in tissue-resident stem cells. In this narrative review article, we discuss the main ion channels of musculoskeletal stem cells, with a focus on calcium-dependent potassium channels, especially on the large conductance BK channel. We review their expression and function in progenitor cell proliferation, differentiation, and migration and highlight gaps in current knowledge on their involvement in musculoskeletal diseases.
Collapse
Affiliation(s)
- Roland Takács
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Patrik Kovács
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Rana Abdelsattar Ebeid
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - János Almássy
- Department of Physiology, Faculty of Medicine, Semmelweis University, H-1428 Budapest, Hungary
| | - János Fodor
- Department of Physiology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - László Ducza
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Richard Barrett-Jolley
- Department of Musculoskeletal Biology, Faculty of Health and Life Sciences, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L69 3GA, UK
| | - Rebecca Lewis
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK
| | - Csaba Matta
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| |
Collapse
|
4
|
Broeders M, van Rooij J, Oussoren E, van Gestel T, Smith C, Kimber S, Verdijk R, Wagenmakers M, van den Hout J, van der Ploeg A, Narcisi R, Pijnappel W. Modeling cartilage pathology in mucopolysaccharidosis VI using iPSCs reveals early dysregulation of chondrogenic and metabolic gene expression. Front Bioeng Biotechnol 2022; 10:949063. [PMID: 36561048 PMCID: PMC9763729 DOI: 10.3389/fbioe.2022.949063] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 11/09/2022] [Indexed: 12/12/2022] Open
Abstract
Mucopolysaccharidosis type VI (MPS VI) is a metabolic disorder caused by disease-associated variants in the Arylsulfatase B (ARSB) gene, resulting in ARSB enzyme deficiency, lysosomal glycosaminoglycan accumulation, and cartilage and bone pathology. The molecular response to MPS VI that results in cartilage pathology in human patients is largely unknown. Here, we generated a disease model to study the early stages of cartilage pathology in MPS VI. We generated iPSCs from four patients and isogenic controls by inserting the ARSB cDNA in the AAVS1 safe harbor locus using CRISPR/Cas9. Using an optimized chondrogenic differentiation protocol, we found Periodic acid-Schiff positive inclusions in hiPSC-derived chondrogenic cells with MPS VI. Genome-wide mRNA expression analysis showed that hiPSC-derived chondrogenic cells with MPS VI downregulated expression of genes involved in TGF-β/BMP signalling, and upregulated expression of inhibitors of the Wnt/β-catenin signalling pathway. Expression of genes involved in apoptosis and growth was upregulated, while expression of genes involved in glycosaminoglycan metabolism was dysregulated in hiPSC-derived chondrogenic cells with MPS VI. These results suggest that human ARSB deficiency in MPS VI causes changes in the transcriptional program underlying the early stages of chondrogenic differentiation and metabolism.
Collapse
Affiliation(s)
- M. Broeders
- Department of Pediatrics, Erasmus MC University Medical Center, Rotterdam, Netherlands
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, Netherlands
- Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Jgj van Rooij
- Department of Internal Medicine, Erasmus MC Medical Center, Rotterdam, Netherlands
| | - E. Oussoren
- Department of Pediatrics, Erasmus MC University Medical Center, Rotterdam, Netherlands
- Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Tjm van Gestel
- Department of Pediatrics, Erasmus MC University Medical Center, Rotterdam, Netherlands
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, Netherlands
- Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Ca Smith
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Sj Kimber
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Rm Verdijk
- Department of Pathology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Maem Wagenmakers
- Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, Netherlands
- Department of Internal Medicine, Erasmus MC Medical Center, Rotterdam, Netherlands
| | - Jmp van den Hout
- Department of Pediatrics, Erasmus MC University Medical Center, Rotterdam, Netherlands
- Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - At van der Ploeg
- Department of Pediatrics, Erasmus MC University Medical Center, Rotterdam, Netherlands
- Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - R. Narcisi
- Department of Orthopaedics and Sports Medicine, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Wwmp Pijnappel
- Department of Pediatrics, Erasmus MC University Medical Center, Rotterdam, Netherlands
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, Netherlands
- Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
5
|
Li L, Jasmer KJ, Camden JM, Woods LT, Martin AL, Yang Y, Layton M, Petris MJ, Baker OJ, Weisman GA, Petris CK. Early Dry Eye Disease Onset in a NOD.H-2h4 Mouse Model of Sjögren's Syndrome. Invest Ophthalmol Vis Sci 2022; 63:18. [PMID: 35727180 PMCID: PMC9233292 DOI: 10.1167/iovs.63.6.18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Purpose To develop a mouse model of human dry eye disease (DED) for investigation of sex differences in autoimmune-associated dry eye pathology. Methods Ocular surface disease was assessed by quantifying corneal epithelial damage with lissamine green stain in the NOD.H-2h4,IFNγ−/−,CD28−/− (NOD.H-2h4 DKO) mouse model of Sjögren's syndrome (SS). Lacrimal gland function was assessed by tear volume quantification with phenol red thread and lacrimal gland inflammation (i.e., dacryoadenitis) was assessed by quantification of immune cell foci, flow cytometric analysis of immune cell composition, and expression of proinflammatory markers. Results The NOD.H-2h4 DKO mouse model of SS exhibits greater age-dependent increases in corneal damage than in NOD.H-2h4 parental mice and demonstrates an earlier disease onset in females compared to males. The severity of ocular surface disease correlates with loss of goblet cell density, increased conjunctivitis, and dacryoadenitis that is more pronounced in NOD.H-2h4 DKO than NOD.H-2h4 mice. B cells dominate lacrimal infiltrates in 16-week-old NOD.H-2h4 and NOD.H-2h4 DKO mice, but T helper cells and macrophages are also present. Lacrimal gland expression of proinflammatory genes, including the P2X7 and P2Y2 purinergic receptors, is greater in NOD.H-2h4 DKO than NOD.H-2h4 mice and correlates with dacryoadenitis. Conclusions Our results demonstrate for the first time that autoimmune dry eye disease occurs in both sexes of NOD.H-2h4 DKO and NOD.H-2h4 mice, with earlier onset in female NOD.H-2h4 DKO mice when compared to males of the same strain. This study demonstrates that both NOD.H-2h4 and NOD.H-2h4 DKO mice are novel models that closely resemble SS-related and sex-dependent DED.
Collapse
Affiliation(s)
- Lili Li
- Division of Biochemistry, University of Missouri, Columbia, Missouri, United States.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States.,Visual Science and Optometry Center, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Kimberly J Jasmer
- Division of Biochemistry, University of Missouri, Columbia, Missouri, United States.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States
| | - Jean M Camden
- Division of Biochemistry, University of Missouri, Columbia, Missouri, United States.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States
| | - Lucas T Woods
- Division of Biochemistry, University of Missouri, Columbia, Missouri, United States.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States
| | - Adam L Martin
- Division of Biochemistry, University of Missouri, Columbia, Missouri, United States.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States
| | - Yong Yang
- Division of Biochemistry, University of Missouri, Columbia, Missouri, United States.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States.,Department of Ophthalmology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Maria Layton
- Division of Biochemistry, University of Missouri, Columbia, Missouri, United States.,Division of Biological Sciences, University of Missouri, Columbia, Missouri, United States
| | - Michael J Petris
- Division of Biochemistry, University of Missouri, Columbia, Missouri, United States.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States.,Department of Ophthalmology, University of Missouri, Columbia, Missouri, United States
| | - Olga J Baker
- Division of Biochemistry, University of Missouri, Columbia, Missouri, United States.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States.,Department of Otolaryngology-Head and Neck Surgery, University of Missouri, Columbia, Missouri, United States
| | - Gary A Weisman
- Division of Biochemistry, University of Missouri, Columbia, Missouri, United States.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States
| | - Carisa K Petris
- Department of Ophthalmology, University of Missouri, Columbia, Missouri, United States.,Mason Eye Institute, University of Missouri, Columbia, Missouri, United States
| |
Collapse
|
6
|
Zhang K, Wang L, Liu Z, Geng B, Teng Y, Liu X, Yi Q, Yu D, Chen X, Zhao D, Xia Y. Mechanosensory and mechanotransductive processes mediated by ion channels in articular chondrocytes: Potential therapeutic targets for osteoarthritis. Channels (Austin) 2021; 15:339-359. [PMID: 33775217 PMCID: PMC8018402 DOI: 10.1080/19336950.2021.1903184] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 02/06/2023] Open
Abstract
Articular cartilage consists of an extracellular matrix including many proteins as well as embedded chondrocytes. Articular cartilage formation and function are influenced by mechanical forces. Hind limb unloading or simulated microgravity causes articular cartilage loss, suggesting the importance of the healthy mechanical environment in articular cartilage homeostasis and implying a significant role of appropriate mechanical stimulation in articular cartilage degeneration. Mechanosensitive ion channels participate in regulating the metabolism of articular chondrocytes, including matrix protein production and extracellular matrix synthesis. Mechanical stimuli, including fluid shear stress, stretch, compression and cell swelling and decreased mechanical conditions (such as simulated microgravity) can alter the membrane potential and regulate the metabolism of articular chondrocytes via transmembrane ion channel-induced ionic fluxes. This process includes Ca2+ influx and the resulting mobilization of Ca2+ that is due to massive released Ca2+ from stores, intracellular cation efflux and extracellular cation influx. This review brings together published information on mechanosensitive ion channels, such as stretch-activated channels (SACs), voltage-gated Ca2+ channels (VGCCs), large conductance Ca2+-activated K+ channels (BKCa channels), Ca2+-activated K+ channels (SKCa channels), voltage-activated H+ channels (VAHCs), acid sensing ion channels (ASICs), transient receptor potential (TRP) family channels, and piezo1/2 channels. Data based on epithelial sodium channels (ENaCs), purinergic receptors and N-methyl-d-aspartate (NMDA) receptors are also included. These channels mediate mechanoelectrical physiological processes essential for converting physical force signals into biological signals. The primary channel-mediated effects and signaling pathways regulated by these mechanosensitive ion channels can influence the progression of osteoarthritis during the mechanosensory and mechanoadaptive process of articular chondrocytes.
Collapse
Affiliation(s)
- Kun Zhang
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| | - Lifu Wang
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| | - Zhongcheng Liu
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| | - Bin Geng
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| | - Yuanjun Teng
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| | - Xuening Liu
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| | - Qiong Yi
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| | - Dechen Yu
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| | - Xiangyi Chen
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| | - Dacheng Zhao
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| | - Yayi Xia
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| |
Collapse
|
7
|
Dielectrophoresis as a Tool to Reveal the Potential Role of Ion Channels and Early Electrophysiological Changes in Osteoarthritis. MICROMACHINES 2021; 12:mi12080949. [PMID: 34442571 PMCID: PMC8402151 DOI: 10.3390/mi12080949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 07/30/2021] [Accepted: 08/08/2021] [Indexed: 11/16/2022]
Abstract
Diseases such as osteoarthritis (OA) are commonly characterized at the molecular scale by gene expression and subsequent protein production; likewise, the effects of pharmaceutical interventions are typically characterized by the effects of molecular interactions. However, these phenomena are usually preceded by numerous precursor steps, many of which involve significant ion influx or efflux. As a consequence, rapid assessment of cell electrophysiology could play a significant role in unravelling the mechanisms underlying drug interactions and progression of diseases, such as OA. In this study, we used dielectrophoresis (DEP), a technique that allows rapid, label-free determination of the dielectric parameters to assess the role of potassium ions on the dielectric characteristics of chondrocytes, and to investigate the electrophysiological differences between healthy chondrocytes and those from an in vitro arthritic disease model. Our results showed that DEP was able to detect a significant decrease in membrane conductance (6191 ± 738 vs. 8571 ± 1010 S/m2), membrane capacitance (10.3 ± 1.47 vs. 14.5 ± 0.01 mF/m2), and whole cell capacitance (5.4 ± 0.7 vs. 7.5 ± 0.3 pF) following inhibition of potassium channels using 10 mM tetraethyl ammonium, compared to untreated healthy chondrocytes. Moreover, cells from the OA model had a different response to DEP force in comparison to healthy cells; this was seen in terms of both a decreased membrane conductivity (782 S/m2 vs. 1139 S/m2) and a higher whole cell capacitance (9.58 ± 3.4 vs. 3.7 ± 1.3 pF). The results show that DEP offers a high throughput method, capable of detecting changes in membrane electrophysiological properties and differences between disease states.
Collapse
|
8
|
Matta C, Lewis R, Fellows C, Diszhazi G, Almassy J, Miosge N, Dixon J, Uribe MC, May S, Poliska S, Barrett-Jolley R, Fodor J, Szentesi P, Hajdú T, Keller-Pinter A, Henslee E, Labeed FH, Hughes MP, Mobasheri A. Transcriptome-based screening of ion channels and transporters in a migratory chondroprogenitor cell line isolated from late-stage osteoarthritic cartilage. J Cell Physiol 2021; 236:7421-7439. [PMID: 34008188 DOI: 10.1002/jcp.30413] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 04/28/2021] [Accepted: 05/04/2021] [Indexed: 12/13/2022]
Abstract
Chondrogenic progenitor cells (CPCs) may be used as an alternative source of cells with potentially superior chondrogenic potential compared to mesenchymal stem cells (MSCs), and could be exploited for future regenerative therapies targeting articular cartilage in degenerative diseases such as osteoarthritis (OA). In this study, we hypothesised that CPCs derived from OA cartilage may be characterised by a distinct channelome. First, a global transcriptomic analysis using Affymetrix microarrays was performed. We studied the profiles of those ion channels and transporter families that may be relevant to chondroprogenitor cell physiology. Following validation of the microarray data with quantitative reverse transcription-polymerase chain reaction, we examined the role of calcium-dependent potassium channels in CPCs and observed functional large-conductance calcium-activated potassium (BK) channels involved in the maintenance of the chondroprogenitor phenotype. In line with our very recent results, we found that the KCNMA1 gene was upregulated in CPCs and observed currents that could be attributed to the BK channel. The BK channel inhibitor paxilline significantly inhibited proliferation, increased the expression of the osteogenic transcription factor RUNX2, enhanced the migration parameters, and completely abolished spontaneous Ca2+ events in CPCs. Through characterisation of their channelome we demonstrate that CPCs are a distinct cell population but are highly similar to MSCs in many respects. This study adds key mechanistic data to the in-depth characterisation of CPCs and their phenotype in the context of cartilage regeneration.
Collapse
Affiliation(s)
- Csaba Matta
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Department of Veterinary Preclinical Sciences, School of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Rebecca Lewis
- Department of Veterinary Preclinical Sciences, School of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Christopher Fellows
- Department of Veterinary Preclinical Sciences, School of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Gyula Diszhazi
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Janos Almassy
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Nicolai Miosge
- Department of Prosthodontics, Tissue Regeneration Work Group, Georg August University, Göttingen, Germany
| | - James Dixon
- Wolfson Centre for Stem Cells, Tissue Engineering and Modelling, Centre of Biomolecular Sciences, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Marcos C Uribe
- The Nottingham Arabidopsis Stock Centre (NASC), School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, UK
| | - Sean May
- The Nottingham Arabidopsis Stock Centre (NASC), School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, UK
| | - Szilard Poliska
- Genomic Medicine and Bioinformatic Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Richard Barrett-Jolley
- Department of Musculoskeletal Biology, Faculty of Health and Life Sciences, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - Janos Fodor
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Peter Szentesi
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tibor Hajdú
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Aniko Keller-Pinter
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Erin Henslee
- Department of Mechanical Engineering Sciences, Centre for Biomedical Engineering, University of Surrey, Guildford, UK
| | - Fatima H Labeed
- Department of Mechanical Engineering Sciences, Centre for Biomedical Engineering, University of Surrey, Guildford, UK
| | - Michael P Hughes
- Department of Mechanical Engineering Sciences, Centre for Biomedical Engineering, University of Surrey, Guildford, UK
| | - Ali Mobasheri
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania.,Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland.,Departments of Orthopedics, Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Joint Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
9
|
Fan Y, Gao D, Zhang Y, Zhu J, Zhang F, Wang L, Wen Y, Guo X, Sun S. Genome-Wide Differentially Methylated Region Analysis to Reveal Epigenetic Differences of Articular Cartilage in Kashin-Beck Disease and Osteoarthritis. Front Cell Dev Biol 2021; 9:636291. [PMID: 33732704 PMCID: PMC7957013 DOI: 10.3389/fcell.2021.636291] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 01/26/2021] [Indexed: 12/21/2022] Open
Abstract
Kashin-Beck disease (KBD) is a degenerative osteoarticular disorder, and displays the significant differences with osteoarthritis (OA) regarding the etiology and molecular changes in articular cartilage. However, the underlying dysfunctions of molecular mechanisms in KBD and OA remain unclear. Here, we primarily performed the various genome-wide differential methylation analyses to reveal the distinct differentially methylated regions (DMRs) in conjunction with corresponding differentially methylated genes (DMGs), and enriched functional pathways in KBD and OA. We identified a total of 131 DMRs in KBD vs. Control, and 58 DMRs in OA vs. Controls, and the results demonstrate that many interesting DMRs are linked to DMGs, such as SMOC2 and HOXD3, which are all key genes to regulate cartilage/skeletal physiologic and pathologic process, and are further enriched in skeletal system and limb-associated pathways. Our DMR analysis indicates that KBD-associated DMRs has higher proportion than OA-associated DMRs in gene body regions. KBD-associated DMGs were enriched in wounding and coagulation-related functional pathways that may be stimulated by trace elements. The identified molecular features provide novel clues for understanding the pathogenetic and therapeutic studies of both KBD and OA.
Collapse
Affiliation(s)
- Yue Fan
- School of Public Health, Health Science Center of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission and Collaborative Innovation Center of Endemic Diseases and Health Promotion in Silk Road Region, Xi'an, China
| | - Dalong Gao
- Department of Orthopaedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Orthopaedics, The Central Hospital of Xianyang, Xianyang, China
| | - Yingang Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jiaqiang Zhu
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, United States
| | - Feng Zhang
- School of Public Health, Health Science Center of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission and Collaborative Innovation Center of Endemic Diseases and Health Promotion in Silk Road Region, Xi'an, China
| | - Lu Wang
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yan Wen
- School of Public Health, Health Science Center of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission and Collaborative Innovation Center of Endemic Diseases and Health Promotion in Silk Road Region, Xi'an, China
| | - Xiong Guo
- School of Public Health, Health Science Center of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission and Collaborative Innovation Center of Endemic Diseases and Health Promotion in Silk Road Region, Xi'an, China
| | - Shiquan Sun
- School of Public Health, Health Science Center of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission and Collaborative Innovation Center of Endemic Diseases and Health Promotion in Silk Road Region, Xi'an, China
| |
Collapse
|
10
|
Jeremiasse B, Matta C, Fellows CR, Boocock DJ, Smith JR, Liddell S, Lafeber F, van Spil WE, Mobasheri A. Alterations in the chondrocyte surfaceome in response to pro-inflammatory cytokines. BMC Mol Cell Biol 2020; 21:47. [PMID: 32586320 PMCID: PMC7318434 DOI: 10.1186/s12860-020-00288-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 06/03/2020] [Indexed: 02/07/2023] Open
Abstract
Background Chondrocytes are exposed to an inflammatory micro-environment in the extracellular matrix (ECM) of articular cartilage in joint diseases such as osteoarthritis (OA) and rheumatoid arthritis (RA). In OA, degenerative changes and low-grade inflammation within the joint transform the behaviour and metabolism of chondrocytes, disturb the balance between ECM synthesis and degradation, and alter the osmolality and ionic composition of the micro-environment. We hypothesize that chondrocytes adjust their physiology to the inflammatory microenvironment by modulating the expression of cell surface proteins, collectively referred to as the ‘surfaceome’. Therefore, the aim of this study was to characterize the surfaceome of primary equine chondrocytes isolated from healthy joints following exposure to the pro-inflammatory cytokines interleukin-1-beta (IL-1β) and tumour necrosis factor-alpha (TNF-α). We employed combined methodology that we recently developed for investigating the surfaceome in stem cells. Membrane proteins were isolated using an aminooxy-biotinylation technique and analysed by mass spectrometry using high throughput shotgun proteomics. Selected proteins were validated by western blotting. Results Amongst the 431 unique cell surface proteins identified, a high percentage of low-abundance proteins, such as ion channels, receptors and transporter molecules were detected. Data are available via ProteomeXchange with identifier PXD014773. A high number of proteins exhibited different expression patterns following chondrocyte stimulation with pro-inflammatory cytokines. Low density lipoprotein related protein 1 (LPR-1), thrombospondin-1 (TSP-1), voltage dependent anion channel (VDAC) 1–2 and annexin A1 were considered to be of special interest and were analysed further by western blotting. Conclusions Our results provide, for the first time, a repository for proteomic data on differentially expressed low-abundance membrane proteins on the surface of chondrocytes in response to pro-inflammatory stimuli.
Collapse
Affiliation(s)
- Bernadette Jeremiasse
- Department of Rheumatology & Clinical Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Csaba Matta
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| | - Christopher R Fellows
- Department of Veterinary Pre-Clinical Sciences, School of Veterinary Science and Medicine, University of Surrey, Guildford, UK
| | - David J Boocock
- John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham, NG11 8NS, UK
| | | | | | - Floris Lafeber
- Department of Rheumatology & Clinical Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Willem E van Spil
- Department of Rheumatology & Clinical Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Ali Mobasheri
- Department of Rheumatology & Clinical Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands. .,Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland. .,Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania. .,Centre for Sport, Exercise and Osteoarthritis Research Versus Arthritis, Queen's Medical Centre, Nottingham, UK. .,Department of Orthopedics, UMC Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
11
|
Surface Display Technology for Biosensor Applications: A Review. SENSORS 2020; 20:s20102775. [PMID: 32414189 PMCID: PMC7294428 DOI: 10.3390/s20102775] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/24/2020] [Accepted: 05/11/2020] [Indexed: 02/06/2023]
Abstract
Surface display is a recombinant technology that expresses target proteins on cell membranes and can be applied to almost all types of biological entities from viruses to mammalian cells. This technique has been used for various biotechnical and biomedical applications such as drug screening, biocatalysts, library screening, quantitative assays, and biosensors. In this review, the use of surface display technology in biosensor applications is discussed. In detail, phage display, bacterial surface display of Gram-negative and Gram-positive bacteria, and eukaryotic yeast cell surface display systems are presented. The review describes the advantages of surface display systems for biosensor applications and summarizes the applications of surface displays to biosensors.
Collapse
|
12
|
Yao W, Dai H, Dong P, Gui J. [Differential expression of transient receptor potential vanilloid receptor 4 protein in osteoarthritis and normal cartilages]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2020; 34:63-68. [PMID: 31939237 PMCID: PMC8171821 DOI: 10.7507/1002-1892.201903056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 10/31/2019] [Indexed: 11/03/2022]
Abstract
OBJECTIVE To investigate the differential expression of transient receptor potential vanilloid receptor 4 (TRPV4) protein in the osteoarthritis (OA) and normal cartilages, and explore the role of TRPV4 in the prevention and treatment of OA. METHODS The cartilage tissues from the patients of knee OA (OA group) and femoral neck fracture (control group) were taken. In OA group, there were 6 males and 9 females; the age ranged from 55 to 78 years (mean, 69 years); the Kellgren-Lawrence (K-L) score was 3.0±0.8. In control group, there were 5 males and 10 females; the age ranged from 57 to 91 years (mean, 71 years). There was no significant difference in gender and age between the two groups ( P>0.05). Western blot, real-time fluorescence quantitative PCR, Masson staining, and immunohistochemical staining were used to detect the difference in protein and mRNA expressions of TRPV4 between the OA and normal cartilages. Then the relationship between the K-L score of OA and the rate of TRPV4-positive cells was analyzed. RESULTS The relative expression of TRPV4 protein and mRNA in OA group were 0.454±0.199 and 2.951±1.200, which were higher than those in control group (0.165±0.074, 1.437±0.682). The difference in relative expression of TRPV4 protein was significant ( t=2.718, P=0.026). Histology observation showed that the chondrocytes arranged disorderly in OA group, the structure of extracellular matrix was abnormal, and the cartilage defect reached the deep layer. There were more TRPV4-positive cells in the degenerated tissue, and the rate of TRPV4-positive cells was 37.353%±13.496%. The chondrocytes were arranged well in control group, and the rate of TRPV4-positive cells was only 9.642%±3.284%. There was a significant difference between the two groups ( t=7.491, P=0.000). The rate of TRPV4-positive cells in OA group was positively correlated with the OA K-L score ( r=0.775, P=0.001). CONCLUSION The TRPV4 expression increased in OA cartilages that may contribute to the development of OA.
Collapse
Affiliation(s)
- Wangxiang Yao
- Department of Sports Medicine and Joint Surgery, the Affiliated Nanjing Hospital of Nanjing Medical University, Nanjing Jiangsu, 210000, P.R.China;Department of Orthopaedics, the Affiliated Hangzhou Hospital of Nanjing Medical University, Hangzhou Zhejiang, 310006, P.R.China
| | - Hanhao Dai
- Department of Sports Medicine and Joint Surgery, the Affiliated Nanjing Hospital of Nanjing Medical University, Nanjing Jiangsu, 210000, P.R.China
| | - Peilong Dong
- Department of Sports Medicine and Joint Surgery, the Affiliated Nanjing Hospital of Nanjing Medical University, Nanjing Jiangsu, 210000, P.R.China
| | - Jianchao Gui
- Department of Sports Medicine and Joint Surgery, the Affiliated Nanjing Hospital of Nanjing Medical University, Nanjing Jiangsu, 210000,
| |
Collapse
|
13
|
Yamamura H, Suzuki Y, Imaizumi Y. Physiological and Pathological Functions of Cl - Channels in Chondrocytes. Biol Pharm Bull 2018; 41:1145-1151. [PMID: 30068862 DOI: 10.1248/bpb.b18-00152] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Articular chondrocytes are embedded in the cartilage of diarthrodial joints and responsible for the synthesis and secretion of extracellular matrix. The extracellular matrix mainly contains collagens and proteoglycans, and covers the articular cartilage to protect from mechanical and biochemical stresses. In mammalian chondrocytes, various types of ion channels have been identified: e.g., voltage-dependent K+ channels, Ca2+-activated K+ channels, ATP-sensitive K+ channels, two-pore domain K+ channels, voltage-dependent Ca2+ channels, store-operated Ca2+ channels, epithelial Na+ channels, acid-sensing ion channels, transient receptor potential channels, and mechanosensitive channels. These channels play important roles for the regulation of resting membrane potential, Ca2+ signaling, pH sensing, mechanotransduction, and cell proliferation in articular chondrocytes. In addition to these cation channels, Cl- channels are known to be expressed in mammalian chondrocytes: e.g., voltage-dependent Cl- channels, cystic fibrosis transmembrane conductance regulator channels, swelling-activated Cl- channels, and Ca2+-activated Cl- channels. Although these chondrocyte Cl- channels are thought to contribute to the regulation of resting membrane potential, Ca2+ signaling, cell volume, cell survival, and endochondral bone formation, the physiological functions have not been fully clarified. Osteoarthritis (OA) is caused by the degradation of articular cartilage, resulting in inflammation and pain in the joints. Therefore the pathophysiological roles of Cl- channels in OA chondrocytes are of considerable interest. Elucidating the physiological and pathological functions of chondrocyte Cl- channels will provide us a more comprehensive understanding of chondrocyte functions and may suggest novel molecular targets of drug development for OA.
Collapse
Affiliation(s)
- Hisao Yamamura
- Department of Molecular & Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University
| | - Yoshiaki Suzuki
- Department of Molecular & Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University
| | - Yuji Imaizumi
- Department of Molecular & Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University
| |
Collapse
|