Role of Targeted Protein Degradation in Immunology

Introduction to Protein Degradation

Protein degradation is a normal and crucial part of the multiple regulatory mechanisms to maintain homeostasis within the human body. Proteins are constantly destroyed and renewed through protein synthesis. Different proteins are degraded at different rates, and each cell has multiple systems that selectively carry out proteolysis. Targeting this process provides a therapeutic approach to diseases that target pathways crucial within human immunology.

LDL Receptors

Image Credit: Juan Gaertner/Shutterstock.com

Protein Degradation Pathways

Eukaryotic cells use proteosomes to degrade proteins via the ubiquitin-proteosome pathway (UPP), and lysosomes degrade protein aggregates and whole organelles through endocytosis/phagocytosis. They also use lysosomes to degrade proteins and organelles through endocytosis, phagocytosis, and autophagy.

The main pathway is the UPP which is crucial in human immunology and regulates immune surveillance. The UPP marks proteins for degradation into small peptides, which can be digested into amino acids. The peptides that are not digested are then recognized by the Major Histocompatibility Complex (MHC), a crucial part of the immune system. Once these peptides are taken up by antigen-presenting cells, they are presented to CD4+ lymphocytes on the surface of MHC molecules, resulting in antibody production.

This process can be modified as a targeted mechanism that aids drug development and treatment of pathogenic and autoimmune diseases. This is known as targeted protein degradation and has a major role in human immunology

What is Targeted Protein Degradation (TPD)

First proposed in 1999, TPD is a therapeutic strategy that aims to target proteins that cause diseases. These proteins have been difficult to target because their active sites are hard to access with conventional small molecule inhibitors. It is important to be able to target these proteins as they have key roles in propagating cancers, autoimmune diseases, and neurodegenerative diseases, so targeting them helps modulate the immunology of these diseases and develop more accurate therapeutic approaches. The drug compounds used aim to disrupt homeostatic mechanisms by interfering with the degradation and processing of proteins.

How does TPD interfere with UPP?

The UPP pathway, as mentioned before, is a crucial part of human immunology and an important component of this pathway is the E3 enzyme which recognizes the targeted protein. This interacts with a 26S proteosome which degrades the protein.

TPD approaches that interfere with the UPP pathway target the E3 ligase enzyme family. One of the earliest TPD was developed 20 years ago and is known as proteolysis-targeting chimera (PROTAC) molecules, also known as protein degraders. These molecules are made up of 2 ligands, where one binds the target protein, and the other binds the E3 ligase. This results in the activation of the UPP system and degradation of the target protein. PROTACs were proven effective in a clinical setting in 2019 against the estrogen receptor (ER) and androgen receptor (AR), a common cancer target.

How does TPD interfere with the lysosomal pathway?

The lysosomal pathway mediates an important aspect of immunology by governing the degradation of intracellular proteins.

A TPD strategy known as LYTAC induces the endosome-lysosome pathway to degrade proteins present extracellularly and as part of the membrane. This technique allows LYTAC molecules to bind an extracellular domain of the target protein and a lysosome-targeting receptor (TLR) present on the cell surface. This forms a complex that internalizes the protein and degrades it through endocytosis.

How can TPD help treat viral diseases?

Viruses are known to be a unique challenge to the immunology of the human body. A successful vaccination program and antiviral drugs are known to combat this. However, the rise of resistance against treatment hinders these efforts, requiring the development of new therapeutic strategies.

Many viruses have evolved mechanisms to avoid immune defense, with one such virus being HIV. It encodes the Nef protein, which targets CD4+ molecules and MHC proteins for degradation via the lysosomal pathway. This affects adaptive immunology by interfering with T- cell responses.

HIV

Image Credit: RAJ CREATIONZS/Shutterstock.com

TPD treatment for Cancer

A lot of cancer-causing proteins hinder immunological processes. For example, BCL-XL is a protein that interferes with cell death and causes malignant cells to escape degradation and regulation. Therefore, it can be an important target for TPD. Significant side effects, such as severe platelet deficiency, were observed with small- molecule inhibitors, calling for the development of TPD strategies. Recent research has shown reduced platelet deficiency with a potent PROTAC targeting BCL-XL.

Another example that showcases the therapeutic effects exhibited by PROTAC is ARV-471. This molecule targets the estrogen receptor (ER) protein, which is involved in the development of breast cancer. Through preclinical and clinical trials, it has been established that degradation reduces tumor growth in xenograft models.

TPD has a major role in immunology as it protects crucial mechanisms when pathogens, cancers, and other diseases target the mechanisms that protect and regulate key functions. There has been a major success with the development of new TPD molecules that are promising drug candidates. However, there is still a plethora of research that needs to be carried out to provide conclusive evidence for the effectiveness of TPD.

Continue Reading: Understanding Neurobiology through Proteins

Sources:

  • Békés, M., Langley, D. R. & Crews, C. M. (2022). ‘Protac targeted protein degraders: The past is prologue’ Nature Reviews Drug Discovery, 21 (3), pp. 181-200. DOI: 10.1038/s41573-021-00371-6 Available at: https://doi.org/10.1038/s41573-021-00371-6.
  • Grohmann, C., Marapana, D. S. & Ebert, G. (2022). ‘Targeted protein degradation at the host-pathogen interface’ Mol Microbiol, 117 (3), pp. 670-681.  DOI: 10.1111/mmi.14849.
  • Lin, X., Xiang, H. & Luo, G. (2020). ‘Targeting estrogen receptor α for degradation with protacs: A promising approach to overcome endocrine resistance’ Eur J Med Chem, 206  p. 112689. DOI: 10.1016/j.ejmech.2020.112689.
  • Reinstein, E. (2004). ‘Immunologic aspects of protein degradation by the ubiquitin-proteasome system’ Isr Med Assoc J, 6 (7), pp. 420-4.
  • Xu, J., Brosseau, J.-P. & Shi, H. (2020). ‘Targeted degradation of immune checkpoint proteins: Emerging strategies for cancer immunotherapy’ Oncogene, 39 (48), pp. 7106-7113. DOI: 10.1038/s41388-020-01491-w  Available at: https://doi.org/10.1038/s41388-020-01491-w.
  • Zhao, L., et al. (2022). ‘Targeted protein degradation: Mechanisms, strategies and application’ Signal Transduct Target Ther, 7 (1), p. 113. DOI: 10.1038/s41392-022-00966-4.

Further Reading

Last Updated: Nov 2, 2022

Ikshitaa Dinesh

Written by

Ikshitaa Dinesh

I’m Ikshitaa Dinesh. I obtained my BSc (Hons) in Biomedicine in 2017 from the University of Huddersfield. During my degree, I was able to undertake a placement year at the UK Health Security Agency (UKHSA), Porton Down from 2019-2020. This is where I discovered my passion for scientific research, especially virology. Initially my project involved, investigating the effect of new nebulisers and air samplers on the survival of influenza virus. However, due to the COVID-19 pandemic, I was unable to continue this research, but I was able to partake in UKHSA’s response to COVID-19. I was part of a team, that carried out a series of experiments to assess the environmental and aerosol survival of SARS-CoV-2. This experience culminated, in being part of 2 research publications titled ‘Persistence of SARS-CoV-2 virus and viral RNA on hydrophobic and hydrophilic surfaces and investigating contamination concentration’ and ‘Characterisation of Particle Size and Viability of SARS-CoV-2 Aerosols from a Range of Nebuliser Types Using a Novel Sampling Technique’ in the journals, Applied and Environmental Microbiology and Viruses, an open access journal published by MDPI, respectively. Here, I was credited for investigation, data acquisition, methodology.

Citations

Please use one of the following formats to cite this article in your essay, paper or report:

  • APA

    Dinesh, Ikshitaa. (2022, November 02). Role of Targeted Protein Degradation in Immunology. AZoLifeSciences. Retrieved on May 11, 2024 from https://www.azolifesciences.com/article/Role-of-Targeted-Protein-Degradation-in-Immunology.aspx.

  • MLA

    Dinesh, Ikshitaa. "Role of Targeted Protein Degradation in Immunology". AZoLifeSciences. 11 May 2024. <https://www.azolifesciences.com/article/Role-of-Targeted-Protein-Degradation-in-Immunology.aspx>.

  • Chicago

    Dinesh, Ikshitaa. "Role of Targeted Protein Degradation in Immunology". AZoLifeSciences. https://www.azolifesciences.com/article/Role-of-Targeted-Protein-Degradation-in-Immunology.aspx. (accessed May 11, 2024).

  • Harvard

    Dinesh, Ikshitaa. 2022. Role of Targeted Protein Degradation in Immunology. AZoLifeSciences, viewed 11 May 2024, https://www.azolifesciences.com/article/Role-of-Targeted-Protein-Degradation-in-Immunology.aspx.

Comments

The opinions expressed here are the views of the writer and do not necessarily reflect the views and opinions of AZoLifeSciences.
Post a new comment
Post

While we only use edited and approved content for Azthena answers, it may on occasions provide incorrect responses. Please confirm any data provided with the related suppliers or authors. We do not provide medical advice, if you search for medical information you must always consult a medical professional before acting on any information provided.

Your questions, but not your email details will be shared with OpenAI and retained for 30 days in accordance with their privacy principles.

Please do not ask questions that use sensitive or confidential information.

Read the full Terms & Conditions.