null
Immunometabolism – A therapeutic perspective

Immunometabolism – A therapeutic perspective

What is Immunometabolism?

Immunometabolism is an exponentially growing, multi-disciplinary field of research aiming at deciphering the dynamic cellular and molecular mechanisms that interweave metabolic and immunological processes together. Although the term “immunometabolism” first appeared in the literature in 2011, the first studies investigating the connection between immune and metabolic disorders date back to the late 19th century. Our expanding understanding of how different immune cell functions correlate with particular metabolic configurations during homeostasis and inflammation has opened new therapeutic possibilities where the modulation of immune responses might be achieved/facilitated by metabolic reprogramming of immune cells.

Explore Immunometabolism assays

T cell metabolism at the steady-state and during an immune response

Among all immune cell subsets currently identified, T lymphocytes are generally considered of particular interest due to their crucial role in anti-tumor, anti-viral and autoimmune responses. At the steady-state, naïve T cells mainly operate under catabolic metabolic programs (oxidative phosphorylation) that maximize energy production (in the form of ATP) over biosynthesis, in order to ensure resting cell survival and basal antigenic immune surveillance. Upon activation (following proper antigenic and co-stimulation cues), T cells must rapidly acquire specific effector functions, change their migratory patterns and actively proliferate – all changes that are highly demanding in energy. The activation of naïve T cells is therefore concomitant with a metabolic reprogramming where oxidative phosphorylation is replaced by the anabolic process of aerobic glycolysis: effector T cells will preferentially ferment glucose to satisfy their energy requirements, even though sufficient oxygen is present to ensure mitochondrial oxidative phosphorylation which would have a better ATP yield (this phenomenon is known as the “Warburg effect”, and is incidentally also observed in cancer cells). Ultimately, once the antigen challenge is cleared, most effector T cells undergo apoptosis, while a pool of long-lasting memory T cells remain to provide enhanced defense against a potential subsequent antigen re-encounter. Memory T cells return to using oxidative pathways to support their long life-span and low turnover rate. Interestingly, memory T cells display a greater spare respiratory capacity in their mitochondria compared to effector T cells, which allows them to rapidly produce ATP in case of secondary antigen exposure, and to quickly and effectively respond to the challenge.

Naïve T cells can differentiate into a variety of subsets. For instance, CD4+ T cells can differentiate into T helper type 1 (characterized by interferon γ secretion), type 2 (IL-4 and IL-13 secretion), type 17 (IL-17 and IL-22 secretion) or regulatory T cells. Importantly, metabolic reprogramming events are not uniform among all T cell subsets and metabolic pathways are central in regulating T cell differentiation and functional responses. Understanding how specific immune functions are endorsed by distinct metabolic programs can help design therapies targeting particular metabolic pathways to precisely modulate T cell differentiation and effector functions. For example, metabolic interventions impacting the ratio between effector and regulatory T cells could help tip the balance between immunogenic and tolerogenic phenotypes, in order to boost or dampen immune responses in specific clinical applications such as cancers or autoimmune diseases.

Main methods used to assess metabolism in immune cells

The methods mentioned below summarize the main experimental assays which can be used (individually or in combination) to characterize metabolic processes in immune cells and their dynamic changes in health and disease.

Measuring glucose uptake:

Glucose entry into cells is one of the early steps necessary to ensure that energy requirements are met, quantifying the rates of glucose uptake can give insight into the metabolic status of immune cells. To perform glucose transport assays, cells are first incubated with a traceable form of glucose: historically, radio-labelled glucose (like 2-deoxy-D-[3H] glucose) was used but is now being replaced by fluorescent glucose analogs (like 2-NBDG or 6-NBDG) that are easier to handle. After a certain period of time, the amount of tracer that has been taken-up by the cells is quantified through scintillation counting or flow cytometry, and then normalized to cell number.

Measuring glucose transporter expression:

In human cells, cell surface and/or intracellular expression of glucose transporters, which are mainly represented by members of the Glut family, can be easily assessed by flow cytometry with the use of fluorochrome-labelled antibodies. However, special care must be taken to confirm the staining specificity and efficiency of the detection antibody used, and changes in glucose transporter expression should always be compared with changes in glucose uptake or any other functional metabolic parameter.

Evaluation of mitochondrial function:

A global picture of overall cellular metabolism can be drawn by assessing mitochondrial oxygen consumption and glycolysis. One of the most straightforward technology to do that is through the live-cell metabolic assay platform developed by Agilent with its line of Seahorse XF Bioanalyzers™ that can measure the real-time oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) of live cells in a multi-well plate, while simultaneously allowing compound addition. The measure of these two parameters, and how they evolve in response to various compounds targeting different mediators of mitochondrial function, can help the experimenter to elucidate the mechanisms involved in metabolic dysregulation.

Measure of metabolite flux via mass spectrometry:

The improvement of mass spectrometry technologies over the years has led to the development of new mass spectrometry-based metabolic assays where the usage and flux individual metabolites can be measured ex vivo with the help of isotopically labeled tracers.

Assessment of metabolic inhibitors:

A broad array of chemical inhibitors, targeting various sections of different metabolic pathways, can be used in combination with the assays described above to study dysregulated metabolic processes in immune cells, and determine how they impact their status and function. The table below provides a non-exhaustive list of small molecules that can be used to probe metabolic reprogramming events in immune cells.

Name Target Metabolic Outcome

2-deoxyglucose

↓glycolysis

3-bromopyruvate

↓glycolysis

Ritonavir

↓glycolysis

Dichloroacetate

↓glycolysis

FX11

↓glycolysis

4-CIN

↓glycolysis

DASA and TEPP46

↓HIF1α

C75

↓fatty acid synthesis

Etomoxir

↓fatty acid oxidation

AICAR

↑fatty acid oxidation

Metformin

AMP kinase

↑fatty acid oxidation

↓Complex I

↓ mitochondrial reactive oxygen species.

Cerulenin

↓fatty acid synthesis

Rotenone

Complex I

↓OXPHOS

BPTES

↓glutaminolysis

Oligomycin

↓OXPHOS

TOFA

↓fatty acid synthesis

UK5099

Pyruvate transporter

↓TCA cycle

Immunometabolism as a therapeutic target

As technologies to unravel immunometabolism continue to progress and enable us to learn more about the metabolic changes associated to specific immune cell polarization, activation and function, it also encourages researchers to consider metabolism in immune cells as a potential therapeutic target for diseases where pathogenesis is driven by dysregulated immune responses. This rational has been fueled by early clinical observations that metabolism-targeting drugs currently used for patients with metabolic disorders (diabetes, dyslipidemia…) also affect immune cell function, thereby opening a way to reposition these drugs as immunomodulators.

We will briefly describe here two clinical settings where immunometabolism has shown potential as a therapeutic target: cancers and autoimmune diseases. In both cases, pathogenesis is attributable to an imbalance between effector and regulatory T cell responses. In cancer, the aim is to enhance beneficial anti-tumoral effector T cell responses and attenuate regulatory processes; while on the contrary in autoimmune disease, the goal is to dampen deleterious overactivated autoreactive effector T cells and boost regulatory responses. For both conditions, an active line of therapeutic research revolves around cell-based immunotherapies that rely on the generation of robust long-lasting populations of antigen-specific T cells whose metabolic pathways can be targeted to enhance their function and persistence in vivo.

Targeting immunometabolism in cancers

A first line of thought when considering the modulation of metabolism in anti-tumoral T cell responses as a therapeutic intervention is to appreciate that the tumoral microenvironment itself (ie hypoxia, nutrient deprivation and local immunosuppression induced by growing cancer cells) profoundly impacts the phenotype and function of T cells in situ. Consequently, metabolism-targeting interventions must not only distinguish between T cells and cancer cells to limit undesirable off-target effects, but immunotherapy products – typically, anti-tumoral T cells isolated from the patient, expanded/modified in vitro and transferred back to the patient – must also be robust enough to resist metabolic changes by the tumoral microenvironment.

The table below recapitulates some of the molecules targeting metabolic pathways that can be used to improve anti-tumoral responses in vivo or as conditioning agents during the in vitro manufacturing of cell-based immunotherapeutic products.

Molecule Target Metabolic Outcome Effects on T Cells

2-deoxy-D-glucose

↓ glycolysis

↑ memory T cell generation.

↑ anti-tumoral T cell function.

Mdivi-1

↓ Mitochondrial fission

↑ anti-tumoral response.

JQ1

↓ glycolysis

↑ T cell longevity and anti-tumor activity.

Rapamycin

↓ glutamine metabolism

↑memory T cell generation.

Metformin

↑ fatty acid oxidation

↑ memory T cell generation.

Fenofibrate

↑ fatty acid catabolism

↑ T cell activity.

Etomoxir

↓ fatty acid oxidation

↓ regulatory T cell differentiation.

1-methyl-tryptophan

↓ tryptophan catabolism

Reactivation of exhausted or tolerant tumoral-specific T cells.

INCB024360

↓ tryptophan catabolism

Reactivation of exhausted or tolerant tumoral-specific T cells.

Ipilimumab

↑ glucose metabolism

↓ CTLA4-mediated T cell inhibition.

Imatinib

TK

↓ glucose uptake

↓ tryptophan catabolism

↓ Lck-mediated T cell receptor signaling.

↑ CD8 T cell activation.

↑ regulatory T cell apoptosis.

Targeting immunometabolism in autoimmune diseases

In autoimmunity, modulating metabolism to limit the activation of autoimmune effector cells and favor the emergence of immune cells with a regulatory phenotype could present an interesting therapeutic approach to control inflammatory autoimmune responses. In that perspective, some encouraging results obtained in experimental models of rheumatic diseases could potentially be translated to the clinic for the treatment of pathologies such as systemic lupus erythematosus, rheumatoid arthritis and osteoarthritis. Interestingly, some standard of care treatments in these diseases already impact metabolic processes (e.g: methotrexate, metformin).

The table below provides a few examples of metabolic drugs that could improve clinical outcome in rheumatic diseases and are currently under investigation.

Drug Target Metabolic outcome Effects on T Cells

Methotrexate

↓ One-carbon metabolism

↓cell growth

Redox balance modulation

Epigenetic modulation

Metformin

↓ glutamine metabolism

↓ cytokine production

↓ B cell differentiation

↓ Th17 differentiation

↑ Treg differentiation

2-deoxy-D-glucose

↓ Glycolysis

↓ proliferation

↓ activation

Bz-423

F-F-ATPase

↓ Mitochondrial oxidation

↑ lymphocyte apoptosis

Dichloroacetate

↓ Glycolysis

↑ ROS

↑ Treg differentiation

3-bromopyruvate

↓ Glycolysis

↓ Th17 differentiation

C968

↓ Glutaminolysis

↓ proliferation

↓ activation

Conclusions

As we garner more knowledge about the close relationship between T cell metabolism and T cell survival, differentiation and functionality, we also come to truly appreciate how deeply interconnected immune networks and systemic metabolism are. Therapeutic approaches aiming at modulating immune responses though metabolism, although promising, often report contradictory results in pre-clinical and clinical studies, and therefore warrant additional investigations to characterize their effects on different immune subsets in various microenvironments.

References

All the information provided above were extracted from the following publications, and the reader is advised to look further into them from a comprehensive and detailed overview of the subject.

Beezhold, K., and Byersdorfer, C.A. (2018). Targeting Immuno-metabolism to Improve Anti-Cancer Therapies. Cancer Lett 414, 127–135.

Chang, C.-H., and Pearce, E.L. (2016). Emerging concepts in immunotherapy – T cell metabolism as a therapeutic target. Nat Immunol 17, 364–368.

Dugnani, E., Pasquale, V., Bordignon, C., Canu, A., Piemonti, L., and Monti, P. (2017). Integrating T cell metabolism in cancer immunotherapy. Cancer Letters 411, 12–18.

Eleftheriadis, T., Pissas, G., Karioti, A., Antoniadi, G., Antoniadis, N., Liakopoulos, V., and Stefanidis, I. (2013). Dichloroacetate at therapeutic concentration alters glucose metabolism and induces regulatory T-cell differentiation in alloreactive human lymphocytes. J Basic Clin Physiol Pharmacol 24, 271–276.

Goropevšek, A., Holcar, M., Pahor, A., and Avčin, T. (2019). STAT signaling as a marker of SLE disease severity and implications for clinical therapy. Autoimmun Rev 18, 144–154.

Hotamisligil, G.S. (2017). Foundations of Immunometabolism and Implications for Metabolic Health and Disease. Immunity 47, 406–420.

Jiang, S., and Yan, W. (2016). T-cell immunometabolism against cancer. Cancer Letters 382, 255–258.

Lee, S.-Y., Moon, S.-J., Kim, E.-K., Seo, H.-B., Yang, E.-J., Son, H.-J., Kim, J.-K., Min, J.-K., Park, S.-H., and Cho, M.-L. (2017). Metformin Suppresses Systemic Autoimmunity in Roquinsan/san Mice through Inhibiting B Cell Differentiation into Plasma Cells via Regulation of AMPK/mTOR/STAT3. J. Immunol. 198, 2661–2670.

Lee, Y.S., Wollam, J., and Olefsky, J.M. (2018). An Integrated View of Immunometabolism. Cell 172, 22–40.

Loftus, R.M., and Finlay, D.K. (2016). Immunometabolism: Cellular Metabolism Turns Immune Regulator. J. Biol. Chem. 291, 1–10.

Mathis, D., and Shoelson, S.E. (2011). Immunometabolism: an emerging frontier. Nat Rev Immunol 11, 81.

Mockler, M.B., Conroy, M.J., and Lysaght, J. (2014). Targeting T Cell Immunometabolism for Cancer Immunotherapy; Understanding the Impact of the Tumor Microenvironment. Front Oncol 4.

Norata, G.D., Caligiuri, G., Chavakis, T., Matarese, G., Netea, M.G., Nicoletti, A., O’Neill, L.A.J., and Marelli-Berg, F.M. (2015). The Cellular and Molecular Basis of Translational Immunometabolism. Immunity 43, 421–434.

Okano, T., Saegusa, J., Nishimura, K., Takahashi, S., Sendo, S., Ueda, Y., and Morinobu, A. (2017). 3-bromopyruvate ameliorate autoimmune arthritis by modulating Th17/Treg cell differentiation and suppressing dendritic cell activation. Sci Rep 7, 42412.

Okano, T., Saegusa, J., Takahashi, S., Ueda, Y., and Morinobu, A. (2018). Immunometabolism in rheumatoid arthritis. Immunological Medicine 41, 89–97.

O’Neill, L.A.J., Kishton, R.J., and Rathmell, J. (2016). A guide to immunometabolism for immunologists. Nat Rev Immunol 16, 553–565.

Palmer, C.S., Henstridge, D.C., Yu, D., Singh, A., Balderson, B., Duette, G., Cherry, C.L., Anzinger, J.J., Ostrowski, M., and Crowe, S.M. (2016). Emerging Role and Characterization of Immunometabolism: Relevance to HIV Pathogenesis, Serious Non-AIDS Events, and a Cure. J Immunol 196, 4437–4444.

Pearce, E.J., and Pearce, E.L. (2018). Driving immunity: all roads lead to metabolism. Nat Rev Immunol 18, 81–82.

Rhoads, J.P., Major, A.S., and Rathmell, J.C. (2017). Fine tuning of immunometabolism for the treatment of rheumatic diseases. Nat Rev Rheumatol 13, 313–320.

Singer, K., Cheng, W.-C., Kreutz, M., Ho, P.-C., and Siska, P.J. (2018). Immunometabolism in cancer at a glance. Dis Model Mech 11.

Starke, I., Glick, G.D., and Börsch, M. (2018). Visualizing Mitochondrial FoF1-ATP Synthase as the Target of the Immunomodulatory Drug Bz-423. Front Physiol 9.

Sukumar, M., Kishton, R.J., and Restifo, N.P. (2017). Metabolic reprograming of anti-tumor immunity. Curr Opin Immunol 46, 14–22.

Sundberg, T.B., Swenson, L., Wahl, D.R., Opipari, A.W., and Glick, G.D. (2009). Apoptotic Signaling Activated by Modulation of the F0F1-ATPase: Implications for Selective Killing of Autoimmune Lymphocytes. J Pharmacol Exp Ther 331, 437–444.

Takahashi, S., Saegusa, J., Sendo, S., Okano, T., Akashi, K., Irino, Y., and Morinobu, A. (2017). Glutaminase 1 plays a key role in the cell growth of fibroblast-like synoviocytes in rheumatoid arthritis. Arthritis Res. Ther. 19, 76.

Takeshima, Y., Iwasaki, Y., Fujio, K., and Yamamoto, K. (2019). Metabolism as a key regulator in the pathogenesis of systemic lupus erythematosus. Seminars in Arthritis and Rheumatism.

Thomas, S., Fisher, K.H., Snowden, J.A., Danson, S.J., Brown, S., and Zeidler, M.P. (2015). Methotrexate Is a JAK/STAT Pathway Inhibitor. PLoS ONE 10, e0130078.

Yin, Y., Choi, S.-C., Xu, Z., Perry, D.J., Seay, H., Croker, B.P., Sobel, E.S., Brusko, T.M., and Morel, L. (2015). Normalization of CD4+ T cell metabolism reverses lupus. Sci Transl Med 7, 274ra18.

Yin, Y., Choi, S.-C., Xu, Z., Zeumer, L., Kanda, N., Croker, B.P., and Morel, L. (2016). Glucose oxidation is critical for CD4+ T cell activation in a mouse model of systemic lupus erythematosus. J Immunol 196, 80–90.

Additional Resources


10th Jul 2023 Dr Celine Van Damme

Recent Posts