Loading

Editorial Open Access
Volume 4 | Issue 1 | DOI: https://doi.org/10.46439/rheumatology.4.025

Exploring novel therapeutic avenues for arthritis precision medicine: The potential of mTOR-SIRT1, NRF2, GPX4 ferroptosis- and autophagy- related pathways and emerging technologies

  • 1Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, Guangdong, China
  • 2Xijing Hospital of Air Force Military Medical University, Xian, China
  • 3Hezhou (the City of Longevity) Dongrong Yao Medicine Research Institute, Joint Institute of Shenzhen University and Hezhou Hospital for Traditional Chinese Medicine, Hezhou, Guangxi, China
  • 4Department of Rheumatology and Immunology, The First Clinical College of Harbin Medical University, Harbin, Heilongjiang, China
  • 5Integrated Chinese and Western Medicine Research Institute, TORAMI Matrix Longevity and Healthcare hub, Zheng He Hospital, Changsha, Hunan, China
  • #Contributed equally
+ Affiliations - Affiliations

*Corresponding Author

Yue Zhang, toronto101@163.com

Received Date: August 07, 2024

Accepted Date: October 03, 2024

Editorial

Arthritis remains a common and debilitating disease affecting millions of people worldwide. Osteoarthritis (OA) and rheumatoid arthritis (RA) are among the most common forms of this chronic, degenerative disease, which is moving toward precision medicine [1-8]. Traditional treatment approaches have primarily focused on symptom management and pain relief, often failing to address the underlying molecular mechanisms that drive the progression of arthritis [9].

However, recent advances in biomedical research have opened new avenues for therapeutic intervention by targeting the key molecular pathways involved in the pathogenesis of arthritis. Emerging evidence suggests that modulation of critical signaling cascades such as mTOR, SIRT1, NRF2 and GPX4 holds great promise for the development of more effective and personalized treatment strategies [4,10].

In addition, the integration of cutting-edge technologies, including microphysiological systems (MPS) [11], artificial intelligence (AI) [12,13], single-cell analysis [3], digital twins [14,15], robotic biotechnology [16-18] and advanced 3D,4D/5D bioprinting [19,20] offers the potential to improve the understanding, diagnosis and treatment of this debilitating disease. By harnessing these innovative approaches, researchers and clinicians can explore new ways to improve the efficacy and precision of arthritis therapies, ultimately contributing to improved patient outcomes and quality of life.

This editorial explores the exciting prospects of combining the modulation of key molecular pathways with the implementation of these transformative technologies in the field of arthritis management. By reviewing recent advances and their potential impact, this article aims to shed light on the promising future of arthritis treatment and the opportunities for revolutionizing arthritis care.

Molecular Pathways and Their Therapeutic Potential

mTOR-SIRT1 pathway

The mTOR (mechanistic target of rapamycin) pathway is a central regulator of cell growth, metabolism, and survival and acts as a sensor of nutrient availability and cellular energy status [8,21]. In arthritis, dysregulated mTOR signaling contributes to increased inflammatory responses and impaired autophagy, exacerbating joint damage [8,22]. SIRT1 (sirtuin 1), an NAD+-dependent deacetylase, counteracts these effects by promoting autophagy and reducing inflammatory cytokine production, thereby increasing cellular resilience [23,24].  

Modulation of the mTOR-SIRT1 axis is a promising therapeutic strategy. For example, the mTOR inhibitor rapamycin has shown potential to reduce cartilage degradation and inflammation in preclinical models of arthritis [8]. Similarly, activation of SIRT1 using small molecules such as resveratrol has demonstrated protective effects against joint degeneration [25].

NRF2 pathway

NRF2 (Nuclear Factor Erythroid 2-Related Factor 2) is a transcription factor that regulates the expression of antioxidant and cytoprotective genes, providing a defense mechanism against oxidative stress. In arthritis, chronic oxidative stress leads to cartilage and synovial tissue damage. Enhancing NRF2 activity could attenuate oxidative damage and inflammation, thus providing therapeutic benefits [26].

Compounds such as sulforaphane, found in cruciferous vegetables, have been shown to activate NRF2 and reduce oxidative stress in arthritis models [27]. In addition, pharmacological NRF2 activators are being investigated for their potential to halt disease progression and improve joint function [28].

GPX4 pathway

GPX4 (glutathione peroxidase 4) is an essential enzyme involved in neutralizing lipid peroxides and preventing ferroptosis, a form of regulated cell death driven by iron-dependent lipid peroxidation [29,30]. In arthritis, ferroptosis contributes to chondrocyte death and cartilage degradation [31]. Increasing GPX4 activity may protect against oxidative damage and maintain cell viability [32].

Strategies to upregulate GPX4 include the use of selenium supplementation, as GPX4 is a selenoprotein, and small molecules that increase GPX4 expression [33]. These approaches hold promise for preserving joint integrity and function.

Innovative Technologies in Arthritis Research

The integration of advanced technologies is revolutionizing arthritis research and therapeutic development.

Microphysiological systems

Microphysiological systems, also known as "organs-on-chips," mimic the microarchitecture and functionality of human tissues in vitro. These systems enable high-throughput screening of drug candidates and provide detailed insights into tissue responses under physiological and pathological conditions [11,34].

For arthritis research, joint-on-a-chip models can mimic the complex interactions between cartilage, synovium and immune cells, allowing precise testing of therapeutic interventions and uncovering disease mechanisms [6,35,36].

Artificial intelligence (AI) and machine learning (ML)

AI and ML are transforming biomedical research by enabling the analysis of large datasets to identify patterns, predict outcomes, and optimize treatment strategies [12]. In arthritis, AI can be used to develop predictive models of disease progression, identify novel drug targets, and personalize treatment regimens based on patient-specific data [13].  For example, AI algorithms have been successfully used to analyze imaging data and quantify joint damage in arthritis patients, improving diagnostic accuracy and monitoring disease progression [37].

Single cell analysis

Single-cell analysis techniques allow the study of cellular heterogeneity within tissues, providing a detailed understanding of cell populations and their role in disease. In arthritis, single-cell RNA sequencing (scRNA-seq) can identify specific cell types and pathways involved in the disease, leading to the development of targeted therapies. Recent studies using scRNA-seq have uncovered previously unrecognized cell types and molecular signatures in arthritic joints, providing new insights into disease mechanisms and potential therapeutic targets [38].

3D, 4D, and 5D bioprinting

3D, 4D, and 5D bioprinting technologies allow for the fabrication of complex, multilayered tissue constructs that mimic the native architecture of human joints [19,39]. These technologies enable the creation of personalized, biocompatible grafts using patient-derived cells, potentially revolutionizing joint repair and regeneration.

For example, 3D bioprinting has been used to create cartilage tissue constructs with precise mechanical properties and cellular composition, showing promise for use in cartilage repair. Advances in 5D bioprinting continue to improve the functionality and realism of printed tissues, paving the way for more effective regenerative medicine [20].

Digital twin technology

Digital twin technology is the creation of a virtual model of a physical entity [15].  In healthcare, this means building a detailed digital replica of a patient's anatomy and pathology that can be used to simulate disease progression and treatment response [14].

Personalized treatment planning: Digital twins equipped with patient-specific data can simulate the effects of different therapeutic interventions, helping physicians choose the most effective treatment plan [40,41].

Predictive modeling: By integrating genomic, proteomic, and clinical data, digital twins can predict disease progression and flare-ups, enabling proactive management of arthritis [42].

Robotic biotechnology

Robotic biotechnology involves the use of robotic systems to perform complex biological tasks with high precision and reproducibility. In the treatment of arthritis, this can revolutionize both surgical and non-surgical interventions. Robotic surgery: Robotic systems can improve the precision of joint surgeries such as arthroplasty or synovectomy, leading to better outcomes and faster recovery times for arthritis patients [16]. Automated drug delivery: Robotic platforms can facilitate targeted drug delivery to inflamed joints, minimizing systemic side effects and maximizing therapeutic efficacy [17]. Finally, nanorobots can also be used [18].

Challenges and Future Directions

While the promising advances in modulating key molecular pathways and integrating innovative technologies have great potential to transform arthritis treatment, several challenges need to be addressed to fully realize the benefits of these approaches.

One of the key challenges is to accurately model the complex human joint environment in vitro [43].  The development of more sophisticated and scalable microphysiological systems will be critical to faithfully recapitulate the cellular and biomechanical interactions within the joint, thereby allowing for more reliable preclinical testing and evaluation of new therapeutic strategies [8,44].

In addition, large-scale validation studies will be needed to confirm the efficacy and safety of emerging molecular and technological interventions [45].  Robust clinical trials incorporating real-world data and multi-omics analyses will be essential to establish the clinical utility of these innovative approaches and ensure their successful translation into routine patient care. In addition, the integration of multidisciplinary expertise spanning fields such as molecular biology, bioengineering, and data science will be critical to the effective implementation of these integrated approaches [46]. Navigating the ethical and regulatory considerations surrounding the use of advanced technologies, such as AI, digital twins and robotic biotechnology, will also be a critical aspect of the future development and deployment of these transformative solutions.

The limitations include lacking accurately modeling joint environments in vitro, the need for more advanced systems, and the necessity of conducting large-scale validation studies. Additionally, improved multidisciplinary collaboration and addressing ethical concerns are critical. The manuscript would also benefit from a discussion on ensuring that patient safety and data privacy remain top priorities.

Despite these limits and challenges, the continued integration of molecular pathway modulation with cutting-edge technologies holds great promise for revolutionizing the treatment of arthritis. By personalizing treatment strategies and increasing the precision of therapeutic interventions, these innovative approaches have the potential to significantly improve the quality of life for patients affected by this debilitating disease.

Future Research Directions Include

Advancing microphysiological systems

Developing more complex and dynamic models that can replicate the multifaceted nature of human joints, including mechanical loading and immune interactions [6,11,13].

Improving AI algorithms

Improving the predictive accuracy of AI models and integrating multi-omics data to provide a comprehensive view of disease mechanisms and treatment responses [47]. The Nobel Prize in Physics 2024 was awarded to both John J. Hopfield and Geoffrey E. Hinton "for foundational discoveries and inventions that enable machine learning with artificial neural networks.

Combining single-cell and multi-omics technologies

Leveraging the power of single-cell analysis in conjunction with genomic, transcriptomic, proteomic, and metabolomic data to achieve a holistic understanding of arthritis [48].

Refining bioprinting techniques

Exploring novel biomaterials and printing methods to improve the durability, functionality, and integration of engineered tissues in joint repair applications [49].

Conclusion

In summary, modulation of key molecular pathways such as mTOR-SIRT1, NRF2, and GPX4, combined with the integration of innovative technologies such as microphysiological systems, artificial intelligence, single-cell analysis, digital twins, robotic biotechnology, and advanced bioprinting, holds great promise for revolutionizing arthritis treatment. These diverse approaches offer new hope for the development of more effective and personalized therapies, ultimately improving the quality of life for patients suffering from this debilitating disease.

The integration of targeted molecular pathway modulation with advanced technologies provides a powerful framework for improving arthritis care. For example, a digital twin model can incorporate patient-specific data on mTOR-SIRT1, NRF2, and GPX4 activity, allowing real-time simulation of how different therapies affect these pathways. Similarly, robotic systems can be programmed to deliver drugs that specifically modulate these molecular targets, improving the precision and efficacy of therapeutic interventions.

The combination of these innovative molecular and technological approaches holds great promise for the future of arthritis treatment. The personalization of treatment strategies and the increased precision of therapeutic interventions have the potential to transform the way arthritis is managed, offering new hope and improved outcomes for patients affected by this debilitating disease.

Acknowledgments of Research Funding

The study was supported by grants from the National Natural Science Foundation of China (No. 81771748), the Shenzhen Science and Technology Project (No. JCYJ20180504170414637), the Futian Healthcare Research Project (No. FTWS2021005), and the Sanming Project of Medicine in Shenzhen (No. SZSM201602087) to Yue Zhang.

References

1. Bhamidipati K, Wei K. Precision medicine in rheumatoid arthritis. Best Pract Res Clin Rheumatol. 2022 Mar;36(1):101742.

2. Li C, Zhang J, Wang W, Wang H, Zhang Y, Zhang Z. Arsenic trioxide improves Treg and Th17 balance by modulating STAT3 in treatment-naïve rheumatoid arthritis patients. Int Immunopharmacol. 2019 Aug;73:539-51.

3. Li C, Chu T, Zhang Z, Zhang Y. Single Cell RNA-Seq Analysis Identifies Differentially Expressed Genes of Treg Cell in Early Treatment-Naive Rheumatoid Arthritis By Arsenic Trioxide. Front Pharmacol. 2021 May 24;12:656124.

4. Dai S, Wang H, Wang M, Zhang Y, Zhang Z, Lin Z. Comparative transcriptomics and network pharmacology analysis to identify the potential mechanism of celastrol against osteoarthritis. Clin Rheumatol. 2021 Oct;40(10):4259-68.

5. Huang C, Zhang Z, Chen Y, Zhang Y, Xing D, Zhao L, et al. Development and formulation of the classification criteria for osteoarthritis. Ann Transl Med. 2020 Sep;8(17):1068.

6. Ye ZZ, Zhang ZY, Li ZG, Huang CB, Zhang Y. Toward wiping out osteoarthritis in China: research highlights. Chin Med J (Engl). 2020 Apr 20;133(8):883-5.

7. Zhang Z, Huang C, Jiang Q, Zheng Y, Liu Y, Liu S, et al. Guidelines for the diagnosis and treatment of osteoarthritis in China (2019 edition). Ann Transl Med. 2020 Oct;8(19):1213.

8. Zhang Y, Chen H, Huang C. Optimizing health-span: advances in stem cell medicine and longevity research. Med Rev (2021). 2023 Oct 10;3(4):351-5.

9. Kolasinski SL, Neogi T, Hochberg MC, Oatis C, Guyatt G, Block J, et al. 2019 American College of Rheumatology/Arthritis Foundation Guideline for the Management of Osteoarthritis of the Hand, Hip, and Knee. Arthritis Care Res (Hoboken). 2020 Feb;72(2):149-62.

10. Dai S, Fan J, Zhang Y, Hao Z, Yu H, Zhang Z. Celastrol promotes chondrocyte autophagy by regulating mTOR expression. Chin Med J (Engl). 2022 Jan 5;135(1):92-4.

11. Ingber DE. Human organs-on-chips for disease modelling, drug development and personalized medicine. Nat Rev Genet. 2022 Aug;23(8):467-91.

12. Esteva A, Robicquet A, Ramsundar B, Kuleshov V, DePristo M, Chou K, et al. A guide to deep learning in healthcare. Nat Med. 2019 Jan;25(1):24-9.

13. Deng S, Li C, Cao J, Cui Z, Du J, Fu Z, et al. Organ-on-a-chip meets artificial intelligence in drug evaluation. Theranostics. 2023 Aug 15;13(13):4526-58.

14. Björnsson B, Borrebaeck C, Elander N, Gasslander T, Gawel DR, Gustafsson M, et al. Swedish Digital Twin Consortium. Digital twins to personalize medicine. Genome Med. 2019 Dec 31;12(1):4.

15. Kamel Boulos MN, Zhang P. Digital Twins: From Personalised Medicine to Precision Public Health. J Pers Med. 2021 Jul 29;11(8):745.

16. Fan H, Marchack K. Biorobotic technologies for regenerative medicine: Current developments and future perspectives. Stem Cell Research & Therapy. 2020;11(1):1-13.

17. Batailler C, Hannouche D, Benazzo F, Parratte S. Concepts and techniques of a new robotically assisted technique for total knee arthroplasty: the ROSA knee system. Arch Orthop Trauma Surg. 2021 Dec;141(12):2049-58.

18. Abaszadeh F, Ashoub MH, Khajouie G, Amiri M. Nanotechnology development in surgical applications: recent trends and developments. Eur J Med Res. 2023 Nov 24;28(1):537.

19. Noroozi R, Arif ZU, Taghvaei H, Khalid MY, Sahbafar H, Hadi A, et al. 3D and 4D Bioprinting Technologies: A Game Changer for the Biomedical Sector? Ann Biomed Eng. 2023 Aug;51(8):1683-712.

20. Tomaskovic-Crook E, Gu Q, Rahim SNA, Wallace GG, Crook JM. Conducting Polymer Mediated Electrical Stimulation Induces Multilineage Differentiation with Robust Neuronal Fate Determination of Human Induced Pluripotent Stem Cells. Cells. 2020 Mar 9;9(3):658.

21. Laplante M, Sabatini DM. mTOR signaling in growth control and disease. Cell. 2012 Apr 13;149(2):274-93.

22. Miller RE, Tran PB, Das R, Ghoreishi-Haack N, Ren D, Miller RJ, et al. CCR2 chemokine receptor signaling mediates pain in experimental osteoarthritis. Proc Natl Acad Sci U S A. 2012 Dec 11;109(50):20602-7.

23. Hubbard BP, Sinclair DA. Small molecule SIRT1 activators for the treatment of aging and age-related diseases. Trends Pharmacol Sci. 2014 Mar;35(3):146-54.

24. Chang N, Li J, Lin S, Zhang J, Zeng W, Ma G, et al. Emerging roles of SIRT1 activator, SRT2104, in disease treatment. Sci Rep. 2024 Mar 6;14(1):5521.

25. El-Bidawy MH, Omar Hussain AB, Al-Ghamdi S, Aldossari KK, Haidara MA, Al-Ani B. Resveratrol ameliorates type 2 diabetes mellitus-induced alterations to the knee joint articular cartilage ultrastructure in rats. Ultrastruct Pathol. 2021 Mar 4;45(2):92-101.

26. Ruan Q, Wang C, Zhang Y, Sun J. Brevilin A attenuates cartilage destruction in osteoarthritis mouse model by inhibiting inflammation and ferroptosis via SIRT1/Nrf2/GPX4 signaling pathway. Int Immunopharmacol. 2023 Nov;124(Pt B):110924.

27. Moon SJ, Jhun J, Ryu J, Kwon JY, Kim SY, Jung K, et al. The anti-arthritis effect of sulforaphane, an activator of Nrf2, is associated with inhibition of both B cell differentiation and the production of inflammatory cytokines. PLoS One. 2021 Feb 16;16(2):e0245986.

28. Shahcheraghi SH, Salemi F, Peirovi N, Ayatollahi J, Alam W, Khan H, et al. Nrf2 Regulation by Curcumin: Molecular Aspects for Therapeutic Prospects. Molecules. 2021 Dec 28;27(1):167.

29. Yang WS, SriRamaratnam R, Welsch ME, Shimada K, Skouta R, Viswanathan VS, et al. Regulation of ferroptotic cancer cell death by GPX4. Cell. 2014 Jan 16;156(1-2):317-31.

30. Zhao H, Tang C, Wang M, Zhao H, Zhu Y. Ferroptosis as an emerging target in rheumatoid arthritis. Front Immunol. 2023 Oct 19;14:1260839.

31. Zhou Y, Que KT, Zhang Z, Yi ZJ, Zhao PX, You Y, et al. Iron overloaded polarizes macrophage to proinflammation phenotype through ROS/acetyl-p53 pathway. Cancer Med. 2018 Aug;7(8):4012-22.

32. Guan Z, Jin X, Guan Z, Liu S, Tao K, Luo L. The gut microbiota metabolite capsiate regulate SLC2A1 expression by targeting HIF-1α to inhibit knee osteoarthritis-induced ferroptosis. Aging Cell. 2023 Jun;22(6):e13807.

33. Ingold I, Berndt C, Schmitt S, Doll S, Poschmann G, Buday K, et al. Selenium Utilization by GPX4 Is Required to Prevent Hydroperoxide-Induced Ferroptosis. Cell. 2018 Jan 25;172(3):409-22.e21.

34. Bhatia SN, Ingber DE. Microfluidic organs-on-chips. Nat Biotechnol. 2014 Aug;32(8):760-72.

35. Ranga A, Gjorevski N, Lutolf MP. Drug discovery through stem cell-based organoid models. Adv Drug Deliv Rev. 2014 Apr;69-70:19-28.

36. Wei K, Korsunsky I, Marshall JL, Gao A, Watts GFM, Major T, et al. Accelerating Medicines Partnership Rheumatoid Arthritis & Systemic Lupus Erythematosus (AMP RA/SLE) Consortium; Siebel CW, Buckley CD, Raychaudhuri S, Brenner MB. Notch signalling drives synovial fibroblast identity and arthritis pathology. Nature. 2020 Jun;582(7811):259-64.

37. Hamet P, Tremblay J. Artificial intelligence in medicine. Metabolism: Clinical and Rxperimental. 2017;69S:S36-S40.

38. Stephenson W, Donlin LT, Butler A, Rozo C, Bracken B, Rashidfarrokhi A, et al. Single-cell RNA-seq of rheumatoid arthritis synovial tissue using low-cost microfluidic instrumentation. Nat Commun. 2018 Feb 23;9(1):791.

39. Murphy SV, Atala A. 3D bioprinting of tissues and organs. Nat Biotechnol. 2014 Aug;32(8):773-85.

40. Bruynseels K, Santoni de Sio F, van den Hoven J. Digital Twins in Health Care: Ethical Implications of an Emerging Engineering Paradigm. Front Genet. 2018 Feb 13;9:31.

41. Padoan A, Plebani M. Dynamic mirroring: unveiling the role of digital twins, artificial intelligence and synthetic data for personalized medicine in laboratory medicine. Clin Chem Lab Med. 2024 May 13.

42. Kozak K, Seidel A, Matvieieva N, Neupetsch C, Teicher U, Lemme G, et al. Unique Device Identification-Based Linkage of Hierarchically Accessible Data Domains in Prospective Surgical Hospital Data Ecosystems: User-Centered Design Approach. JMIR Med Inform. 2023 Jan 27;11:e41614.

43. Johnson PA, Ackerman JE, Kurowska-Stolarska M, Coles M, Buckley CD, Dakin SG. Three-dimensional, in-vitro approaches for modelling soft-tissue joint diseases. Lancet Rheumatol. 2023 Sep;5(9):e553-63.

44. Paggi CA, Teixeira LM, Le Gac S, Karperien M. Joint-on-chip platforms: entering a new era of in vitro models for arthritis. Nat Rev Rheumatol. 2022 Apr;18(4):217-31.

45. Baden LR, El Sahly HM, Essink B, Kotloff K, Frey S, Novak R, et al. COVE Study Group. Efficacy and Safety of the mRNA-1273 SARS-CoV-2 Vaccine. N Engl J Med. 2021 Feb 4;384(5):403-16.

46. Liu R, Rizzo S, Whipple S, Pal N, Pineda AL, Lu M, Arnieri B, Lu Y, Capra W, Copping R, Zou J. Evaluating eligibility criteria of oncology trials using real-world data and AI. Nature. 2021 Apr;592(7855):629-33.

47. Alipanahi B, Delong A, Weirauch MT, Frey BJ. Predicting the sequence specificities of DNA- and RNA-binding proteins by deep learning. Nat Biotechnol. 2015 Aug;33(8):831-8.

48. Rockel S, Sharma D, Espin-Garcia, Hueniken K, Sandhu A, Pastrello C, et al. Deep Learning-Based Multimodal Clustering Model for Endotyping and Post-Arthroplasty Response Classification using Knee Osteoarthritis Subject-Matched Multi-Omic Data. June 2024 Preprint https://doi.org/10.1101/2024.06.13.24308857.

49. Yang J, Jia C, Yang J. Designing Nanoparticle-based Drug Delivery Systems for Precision Medicine. Int J Med Sci. 2021 Jun 5;18(13):2943-9.

Author Information X