Next Article in Journal
A Review on Metal Oxide Semiconductor-Based Chemo-Resistive Ethylene Sensors for Agricultural Applications
Previous Article in Journal
Prostate Cancer Detection in Colombian Patients through E-Senses Devices in Exhaled Breath and Urine Samples
Previous Article in Special Issue
MicroRNA Biosensors for Early Detection of Hepatocellular Carcinoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Advanced Techniques for the Analysis of Proteins and RNAs

Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada
Chemosensors 2024, 12(1), 12; https://doi.org/10.3390/chemosensors12010012
Submission received: 2 January 2024 / Accepted: 8 January 2024 / Published: 10 January 2024
(This article belongs to the Special Issue Advanced Techniques for the Analysis of Protein and RNA)
Proteins and RNAs, as fundamental components of cellular machinery, play pivotal roles in the intricate landscape of life [1,2]. They have a spectrum of functions, ranging from acting as crucial structural elements to orchestrating complex signaling pathways and cellular processes [3,4,5,6,7,8,9,10]. Throughout the dynamic stages of development and in biological responses to environmental factors, proteins and RNAs contribute significantly to the regulation of growth, differentiation, the immune response, and other essential biological processes [5,11,12,13,14,15,16,17,18,19,20,21,22]. Recognizing their multifaceted roles and enabling the rapid and accurate detection of proteins and RNAs, including mRNA and microRNA, are imperative to advancing our understanding of cellular dynamics and their implications in health and disease [11,23,24,25,26,27,28].
The advent of novel analytical techniques in recent years has led to an era of remarkable progress in this field. Technologies such as mass spectrometry [29,30,31,32], immunoassays [33,34,35,36], spectral analysis [37,38], chemosensors [39,40,41], and biosensors [41,42,43,44,45,46,47] have emerged as powerful tools that provide unprecedented insights into the functions and interactions of proteins and RNAs [48,49]. These and many other emerging technological advancements [50,51,52] have not only enhanced our ability to unravel the intricacies of cellular mechanisms, but also hold immense potential to reshape the landscape of healthcare and environmental science. The application of these cutting-edge techniques enables us to take a transformative approach to the diagnosis, monitoring, and treatment of a myriad of diseases, ultimately promising to elevate the quality of life of millions worldwide.
With the advancement of technologies, progress has recently been made in the analysis of proteins and RNAs. In this Special Issue of Chemosensors on “Advanced Techniques for the Analysis of Proteins and RNAs”, I am pleased to present a comprehensive overview of the impactful research showcased in nine published papers. These studies represent cutting-edge methodologies and applications in the fields of biosensing, enzymatic assays, liquid chromatography, and colorimetric and fluorescent assays, providing valuable insights into the detailed analysis of proteins, RNA, and associated pollutants.
Two papers within this collection focus on the development of biosensors tailored for the detection of protein levels within living cells. The work by Wu et al. introduces a novel HiBiT biosensor meticulously designed to monitor the stability of YAP/TAZ proteins in cells [53]. Using this biosensor, the authors identified novel small molecules that regulate YAP/TAZ, offering a promising avenue to improving anti-triple-negative breast cancer (TNBC) therapy. This pioneering approach marks a significant stride in cancer drug discovery through the innovative application of biosensing technologies. In a parallel study, Sitkov et al. engineered an impedimetric biosensor featuring zinc oxide nanorods for the highly sensitive detection of biomarkers [54]. Their work underscores the biosensor’s potential for utilization in point-of-care systems designed for the swift, multimodal detection of molecular markers across diverse diseases.
Several other papers employed peptides or enzymes with a high affinity for proteins to quantify proteins or protein-like reagents in vitro. Zimina et al. dedicated their research to designing a peptide ligand with a heightened affinity for proteins such as lactoferrin, a multifunctional protein derived from milk, employing in silico modeling and capillary electrophoresis techniques to study its binding specificity [55]. Shahzadi et al. explored the feasibility of leveraging ultra-thin 2D materials, renowned for their high affinity for proteins and DNA, for biosensing applications [56]. Their evaluation of the non-covalent functionalization of CVD-graphene using a pyrene-based supporter construct demonstrated its potential in biosensing applications, particularly in the detection of proteins like streptavidin. In their contribution, Esimbekova et al. introduced a trypsin-based chemoenzymatic assay tailored for the detection of pollutants and the safety assessment of food additives [57], showcasing the versatility of enzyme-based detection systems for the analysis of reagents akin to enzymatic proteins.
Additionally, Kuznetsov et al. presented a novel chromatographic sensor designed for fast protein and metabolite liquid chromatography, facilitating the evaluation of fish and meat freshness through the analysis of specific protein and DNA contents [58]. The proposed freshness index (H*) demonstrated a strong correlation with traditional freshness indicators. Moreover, Mamaeva et al. and Li et al. conducted insightful comparisons of various colorimetric and fluorometric chemosensors or assays for protein concentration determination, offering valuable insights into the comparative performance of different dyes and assays [59,60].
Finally, Lin et al. contributed a review article on microRNA biosensors tailored for the early detection of hepatocellular carcinoma, summarizing recent advances in biosensor technologies geared towards detecting these pivotal biomarkers [61].
The diverse range of contributions to this Special Issue collectively advance the field of analytical techniques for protein and RNA analysis, providing novel insights, methodologies, and applications that are poised to catalyze further research in these pivotal areas. I extend my sincere appreciation to all the abovementioned authors for their invaluable contributions to this Special Issue.

Funding

In addition, I would like to thank the Canadian Institute of Health Research (#186143, 119325), Cancer Research Society (#938324), and New Frontier Research Fund (#340043) for their financial support.

Acknowledgments

I thank all the authors who submitted their studies to the present Special Issue.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Alberts, B. The cell as a collection of protein machines: Preparing the next generation of molecular biologists. Cell 1998, 92, 291–294. [Google Scholar] [CrossRef] [PubMed]
  2. Nair, A.; Chauhan, P.; Saha, B.; Kubatzky, K.F. Conceptual Evolution of Cell Signaling. Int. J. Mol. Sci. 2019, 20, 3292. [Google Scholar] [CrossRef] [PubMed]
  3. Ganser, L.R.; Kelly, M.L.; Herschlag, D.; Al-Hashimi, H.M. The roles of structural dynamics in the cellular functions of RNAs. Nat. Rev. Mol. Cell Biol. 2019, 20, 474–489. [Google Scholar] [CrossRef] [PubMed]
  4. Westermarck, J.; Ivaska, J.; Corthals, G.L. Identification of protein interactions involved in cellular signaling. Mol. Cell Proteom. 2013, 12, 1752–1763. [Google Scholar] [CrossRef] [PubMed]
  5. Taha, Z.; Janse van Rensburg, H.J.; Yang, X. The Hippo pathway: Immunity and cancer. Cancers 2018, 10, 94. [Google Scholar] [CrossRef]
  6. Pfleger, C.M. The Hippo Pathway: A Master Regulatory Network Important in Development and Dysregulated in Disease. Curr. Top. Dev. Biol. 2017, 123, 181–228. [Google Scholar]
  7. Ardestani, A.; Lupse, B.; Maedler, K. Hippo Signaling: Key Emerging Pathway in Cellular and Whole-Body Metabolism. Trends Endocrinol. Metab. 2018, 29, 492–509. [Google Scholar] [CrossRef]
  8. Day, E.K.; Sosale, N.G.; Lazzara, M.J. Cell signaling regulation by protein phosphorylation: A multivariate, heterogeneous, and context-dependent process. Curr. Opin. Biotechnol. 2016, 40, 185–192. [Google Scholar] [CrossRef]
  9. Li, X.; Tran, K.M.; Aziz, K.E.; Sorokin, A.V.; Chen, J.; Wang, W. Defining the Protein-Protein Interaction Network of the Human Protein Tyrosine Phosphatase Family. Mol. Cell Proteom. 2016, 15, 3030–3044. [Google Scholar] [CrossRef]
  10. Xu, A.M.; Huang, P.H. Receptor tyrosine kinase coactivation networks in cancer. Cancer Res. 2010, 70, 3857–3860. [Google Scholar] [CrossRef]
  11. Khan, M.G.M.; Wang, Y. Cell Cycle-Related Clinical Applications. Methods Mol. Biol. 2022, 2579, 35–46. [Google Scholar]
  12. Goranov, A.I.; Cook, M.; Ricicova, M.; Ben-Ari, G.; Gonzalez, C.; Hansen, C.; Tyers, M.; Amon, A. The rate of cell growth is governed by cell cycle stage. Genes Dev. 2009, 23, 1408–1422. [Google Scholar] [CrossRef] [PubMed]
  13. Tzur, A.; Kafri, R.; LeBleu, V.S.; Lahav, G.; Kirschner, M.W. Cell growth and size homeostasis in proliferating animal cells. Science 2009, 325, 167–171. [Google Scholar] [CrossRef]
  14. Golubev, A. Transition probability in cell proliferation, stochasticity in cell differentiation, and the restriction point of the cell cycle in one package. Prog. Biophys. Mol. Biol. 2012, 110, 87–96. [Google Scholar] [CrossRef]
  15. Miller, J.P.; Yeh, N.; Vidal, A.; Koff, A. Interweaving the cell cycle machinery with cell differentiation. Cell Cycle 2007, 6, 2932–2938. [Google Scholar] [CrossRef] [PubMed]
  16. Hendriks, D.; Artegiani, B.; Kretzschmar, K. Editorial: Mechanisms of cellular differentiation, organ development, and novel model systems. Front. Cell Dev. Biol. 2022, 10, 970778. [Google Scholar] [CrossRef] [PubMed]
  17. Newman, S.A. Cell differentiation: What have we learned in 50 years? J. Theor. Biol. 2020, 485, 110031. [Google Scholar] [CrossRef]
  18. Yang, X.; Xu, T. Molecular mechanism of size control in development and human diseases. Cell Res. 2011, 21, 715–729. [Google Scholar] [CrossRef]
  19. Nelson, C.M. Mechanical Control of Cell Differentiation: Insights from the Early Embryo. Annu. Rev. Biomed. Eng. 2022, 24, 307–322. [Google Scholar] [CrossRef]
  20. Goddard, A.M.; Cho, M.G.; Lerner, L.M.; Gupta, G.P. Mechanisms of Immune Sensing of DNA Damage. J. Mol. Biol. 2023, 168424. [Google Scholar] [CrossRef]
  21. Chatterjee, N.; Walker, G.C. Mechanisms of DNA damage, repair, and mutagenesis. Environ. Mol. Mutagen. 2017, 58, 235–263. [Google Scholar] [CrossRef]
  22. Yaribeygi, H.; Panahi, Y.; Sahraei, H.; Johnston, T.P.; Sahebkar, A. The impact of stress on body function: A review. Excli. J. 2017, 16, 1057–1072. [Google Scholar] [PubMed]
  23. Ligasová, A.; Frydrych, I.; Koberna, K. Basic Methods of Cell Cycle Analysis. Int. J. Mol. Sci. 2023, 24, 3674. [Google Scholar] [CrossRef] [PubMed]
  24. Roth, S.; Margulis, M.; Danielli, A. Recent Advances in Rapid and Highly Sensitive Detection of Proteins and Specific DNA Sequences Using a Magnetic Modulation Biosensing System. Sensors 2022, 22, 4497. [Google Scholar] [CrossRef] [PubMed]
  25. Duffy, D.C. Short Keynote Paper: Single Molecule Detection of Protein Biomarkers to Define the Continuum from Health to Disease. IEEE J. Biomed. Health Inf. 2020, 24, 1864–1868. [Google Scholar] [CrossRef] [PubMed]
  26. Meng, X.; Pang, X.; Yang, J.; Zhang, X.; Dong, H. Recent Advances in Electrochemiluminescence Biosensors for MicroRNA Detection. Small 2023, e2307701. [Google Scholar] [CrossRef] [PubMed]
  27. Shaterabadi, D.; Zamani Sani, M.; Rahdan, F.; Taghizadeh, M.; Rafiee, M.; Dorosti, N.; Dianatinasab, A.; Taheri-Anganeh, M.; Asadi, P.; Khatami, S.H.; et al. MicroRNA biosensors in lung cancer. Clin. Chim. Acta 2024, 552, 117676. [Google Scholar] [CrossRef] [PubMed]
  28. Quazi, S. Application of biosensors in cancers, an overview. Front. Bioeng. Biotechnol. 2023, 11, 1193493. [Google Scholar] [CrossRef]
  29. Boja, E.S.; Rodriguez, H. Mass spectrometry-based targeted quantitative proteomics: Achieving sensitive and reproducible detection of proteins. Proteomics 2012, 12, 1093–1110. [Google Scholar] [CrossRef]
  30. Shuford, C.M.; Grant, R.P. Cheaper, faster, simpler trypsin digestion for high-throughput targeted protein quantification. J. Mass Spectrom. Adv. Clin. Lab. 2023, 30, 74–82. [Google Scholar] [CrossRef]
  31. Shen, S.; Wang, X.; Zhu, X.; Rasam, S.; Ma, M.; Huo, S.; Qian, S.; Zhang, M.; Qu, M.; Hu, C.; et al. High-quality and robust protein quantification in large clinical/pharmaceutical cohorts with IonStar proteomics investigation. Nat. Protoc. 2023, 18, 700–731. [Google Scholar] [CrossRef] [PubMed]
  32. Popp, R.; Li, H.; Borchers, C.H. Immuno-MALDI (iMALDI) mass spectrometry for the analysis of proteins in signaling pathways. Expert Rev. Proteom. 2018, 15, 701–708. [Google Scholar] [CrossRef] [PubMed]
  33. Shin, H.; Oh, S.; Kang, D.; Choi, Y. Protein Quantification and Imaging by Surface-Enhanced Raman Spectroscopy and Similarity Analysis. Adv. Sci. (Weinh.) 2020, 7, 1903638. [Google Scholar] [CrossRef] [PubMed]
  34. Dixit, C.K.; Kaushik, A. Nano-structured arrays for multiplex analyses and Lab-on-a-Chip applications. Biochem. Biophys. Res. Commun. 2012, 419, 316–320. [Google Scholar] [CrossRef] [PubMed]
  35. Zhou, S.; Lu, X.; Chen, C.; Sun, D. An immunoassay method for quantitative detection of proteins using single antibodies. Anal. Biochem. 2010, 400, 213–218. [Google Scholar] [CrossRef]
  36. Misiewicz-Krzeminska, I.; Corchete, L.A.; Rojas, E.A.; Martínez-López, J.; García-Sanz, R.; Oriol, A.; Bladé, J.; Lahuerta, J.J.; Miguel, J.S.; Mateos, M.V.; et al. A novel nano-immunoassay method for quantification of proteins from CD138-purified myeloma cells: Biological and clinical utility. Haematologica 2018, 103, 880–889. [Google Scholar] [CrossRef]
  37. Gautier, A. Fluorescence-Activating and Absorption-Shifting Tags for Advanced Imaging and Biosensing. Acc. Chem. Res. 2022, 55, 3125–3135. [Google Scholar] [CrossRef]
  38. Fukuyama, M.; Nakamura, A.; Nishiyama, K.; Imai, A.; Tokeshi, M.; Shigemura, K.; Hibara, A. Noncompetitive Fluorescence Polarization Immunoassay for Protein Determination. Anal. Chem. 2020, 92, 14393–14397. [Google Scholar] [CrossRef]
  39. Crisp, S.J.; Dunn, M.J. Detection of proteins on protein blots using chemiluminescent systems. Methods Mol. Biol. 1994, 32, 233–237. [Google Scholar]
  40. Khan, J. Synthesis and Applications of Fluorescent Chemosensors: A Review. J. Fluoresc. 2023. [Google Scholar] [CrossRef]
  41. Xia, N.; Chang, Y.; Zhou, Q.; Ding, S.; Gao, F. An Overview of the Design of Metal-Organic Frameworks-Based Fluorescent Chemosensors and Biosensors. Biosensors 2022, 12, 928. [Google Scholar] [CrossRef] [PubMed]
  42. Kang, M.J.; Cho, Y.W.; Kim, T.H. Progress in Nano-Biosensors for Non-Invasive Monitoring of Stem Cell Differentiation. Biosensors 2023, 13, 501. [Google Scholar] [CrossRef]
  43. Pipchuk, A.; Yang, X. Using Biosensors to Study Protein-Protein Interaction in the Hippo Pathway. Front. Cell Dev. Biol. 2021, 9, 660137. [Google Scholar] [CrossRef] [PubMed]
  44. Azad, T.; Tashakor, A.; Hosseinkhani, S. Split-luciferase complementary assay: Applications, recent developments, and future perspectives. Anal. Bioanal. Chem. 2014, 406, 5541–5560. [Google Scholar] [CrossRef] [PubMed]
  45. Mo, X.; Niu, Q.; Ivanov, A.A.; Tsang, Y.H.; Tang, C.; Shu, C.; Li, Q.; Qian, K.; Wahafu, A.; Doyle, S.P.; et al. Systematic discovery of mutation-directed neo-protein-protein interactions in cancer. Cell 2022, 185, 1974–1985.e12. [Google Scholar] [CrossRef] [PubMed]
  46. Nouri, K.; Azad, T.; Lightbody, E.; Khanal, P.; Nicol, C.J.; Yang, X. A kinome-wide screen using a NanoLuc LATS luminescent biosensor identifies ALK as a novel regulator of the Hippo pathway in tumorigenesis and immune evasion. FASEB J. 2019, 33, 12487–12499. [Google Scholar] [CrossRef] [PubMed]
  47. Azad, T.; Janse van Rensburg, H.J.; Lightbody, E.D.; Neveu, B.; Champagne, A.; Ghaffari, A.; Kay, V.R.; Hao, Y.; Shen, H.; Yeung, B.; et al. A LATS biosensor functional screen identifies VEGFR as a novel regulator of the Hippo pathway in angiogenesis. Nat. Commun. 2018, 9, 1061. [Google Scholar] [CrossRef]
  48. Rao, V.S.; Srinivas, K.; Sujini, G.N.; Kumar, G.N. Protein-protein interaction detection: Methods and analysis. Int. J. Proteom. 2014, 2014, 147648. [Google Scholar] [CrossRef]
  49. Pflieger, D.; Gonnet, F.; de la Fuente van Bentem, S.; Hirt, H.; de la Fuente, A. Linking the proteins--elucidation of proteome-scale networks using mass spectrometry. Mass Spectrom. Rev. 2011, 30, 268–297. [Google Scholar] [CrossRef]
  50. Baysoy, A.; Bai, Z.; Satija, R.; Fan, R. The technological landscape and applications of single-cell multi-omics. Nat. Rev. Mol. Cell Biol. 2023, 24, 695–713. [Google Scholar] [CrossRef]
  51. Hu, Y.; Cheng, K.; He, L.; Zhang, X.; Jiang, B.; Jiang, L.; Li, C.; Wang, G.; Yang, Y.; Liu, M. NMR-Based Methods for Protein Analysis. Anal. Chem. 2021, 93, 1866–1879. [Google Scholar] [CrossRef] [PubMed]
  52. Brunner, A.D.; Thielert, M.; Vasilopoulou, C.; Ammar, C.; Coscia, F.; Mund, A.; Hoerning, O.B.; Bache, N.; Apalategui, A.; Lubeck, M.; et al. Ultra-high sensitivity mass spectrometry quantifies single-cell proteome changes upon perturbation. Mol. Syst. Biol. 2022, 18, e10798. [Google Scholar] [CrossRef] [PubMed]
  53. Wu, L.; Ge, A.; Hao, Y.; Yang, X. Development of a New HiBiT Biosensor Monitoring Stability of YAP/TAZ Proteins in Cells. Chemosensors 2023, 11, 492. [Google Scholar] [CrossRef]
  54. Sitkov, N.; Ryabko, A.; Kolobov, A.; Maximov, A.; Moshnikov, V.; Pshenichnyuk, S.; Komolov, A.; Aleshin, A.; Zimina, T. Impedimetric Biosensor Coated with Zinc Oxide Nanorods Synthesized by a Modification of the Hydrothermal Method for Antibody Detection. Chemosensors 2023, 11, 66. [Google Scholar] [CrossRef]
  55. Zimina, T.; Sitkov, N.; Karasev, V.; Skorik, Y.; Kolobov, A.; Kolobov, A.; Bunenkov, N.; Luchinin, V. Design of Peptide Ligand for Lactoferrin and Study of Its Binding Specificity. Chemosensors 2023, 11, 162. [Google Scholar] [CrossRef]
  56. Shahzadi, M.; Nisar, S.; Kim, D.-K.; Sarwar, N.; Rasheed, A.; Ahmad, W.; Afzal, A.M.; Imran, M.; Assiri, M.A.; Shahzad, Z.M.; et al. Highly Efficient, Non-Covalent Functionalization of CVD-Graphene via Novel Pyrene-Based Supporter Construct. Chemosensors 2023, 11, 83. [Google Scholar] [CrossRef]
  57. Esimbekova, E.N.; Torgashina, I.G.; Nemtseva, E.V.; Antashkevich, A.A.; Sasova, P.Y.; Kratasyuk, V.A. Trypsin-Based Chemoenzymatic Assay for Detection of Pollutants and Safety Assessment of Food Additives. Chemosensors 2023, 11, 237. [Google Scholar] [CrossRef]
  58. Kuznetsov, A.; Frorip, A.; Sünter, A.; Kasvand, N.; Korsakov, V.; Konoplev, G.; Stepanova, O.; Rusalepp, L.; Anton, D.; Püssa, T.; et al. Fast Protein and Metabolites (Nucleotides and Nucleosides) Liquid Chromatography Technique and Chemical Sensor for the Assessment of Fish and Meat Freshness. Chemosensors 2023, 11, 69. [Google Scholar] [CrossRef]
  59. Mamaeva, A.A.; Martynov, V.I.; Deyev, S.M.; Pakhomov, A.A. Comparison of Colorimetric and Fluorometric Chemosensors for Protein Concentration Determination and Approaches for Estimation of Their Limits of Detection. Chemosensors 2022, 10, 542. [Google Scholar] [CrossRef]
  60. Li, F.; Tan, J.; Yang, Q.; He, M.; Yu, R.; Liu, C.; Zhou, X. Multi-Endpoint Toxicity Tests and Effect-Targeting Risk Assessment of Surface Water and Pollution Sources in a Typical Rural Area in the Yellow River Basin, China. Chemosensors 2022, 10, 502. [Google Scholar] [CrossRef]
  61. Lin, X.; Wang, K.; Luo, C.; Yang, M.; Wu, J. MicroRNA Biosensors for Early Detection of Hepatocellular Carcinoma. Chemosensors 2023, 11, 504. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Yang, X. Advanced Techniques for the Analysis of Proteins and RNAs. Chemosensors 2024, 12, 12. https://doi.org/10.3390/chemosensors12010012

AMA Style

Yang X. Advanced Techniques for the Analysis of Proteins and RNAs. Chemosensors. 2024; 12(1):12. https://doi.org/10.3390/chemosensors12010012

Chicago/Turabian Style

Yang, Xiaolong. 2024. "Advanced Techniques for the Analysis of Proteins and RNAs" Chemosensors 12, no. 1: 12. https://doi.org/10.3390/chemosensors12010012

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop