Review
Immunometabolic alterations in lupus: where do they come from and where do we go from there?

https://doi.org/10.1016/j.coi.2022.102245Get rights and content

Highlights

  • Immunometabolism dysregulations in lupus have multifactorial origins.

  • Contributing factors include genetics and chronic autoantigen stimulation.

  • Metabolites from dysbiotic microbiota may regulate immune-cell metabolism.

  • Autoimmune activation and altered cellular metabolism are deeply interconnected.

  • Novel metabolic targets have been identified as potential therapies in lupus.

Systemic lupus erythematosus (SLE) is an autoimmune disease in which the overactivation of the immune system has been associated with metabolic alterations. Targeting the altered immunometabolism has been proposed to treat SLE patients based on their results obtained and mouse models of the disease. Here, we review the recent literature to discuss the possible origins of the alterations in the metabolism of immune cells in lupus, the dominant role of mitochondrial defects, technological advances that may move the field forward, as well as how targeting lupus immunometabolism may have therapeutic potential.

Introduction

Systemic lupus erythematosus (SLE) is a prototypic autoimmune disease driven by pathogenic autoantibodies that results from the activation of multiple immune-cell types. The overactivation of the lupus immune system has been associated with metabolic alterations in SLE patients as well as in mouse models of the disease, which have been recently reviewed 1, 2 Targeting the altered immunometabolism has been proposed to treat SLE patients 3, 4, which has been validated by clinical trials that have shown that sirolimus, a mammalian target of rapamycin (mTOR) inhibitor, reduced disease activity [5], and metformin, a mild inhibitor of complex 1 in the electron-transport chain, reduced flares [6]. Not surprisingly, the field has grown in complexity from the initial findings of increased mTOR activation and glycolysis in CD4+ T cells [1]. Here, we review the recent literature to discuss the possible origins of the alterations in the metabolism of immune cells in lupus (Figure 1), the dominant role of mitochondrial defects (Figure 2), technological advances that may move the field forward (Figure 3), as well as how targeting lupus immunometabolism may have therapeutic potential (Figure 4).

Section snippets

Genetic factors directly altering immune-cell metabolism

Lupus and many of its endophenotypes have a strong genetic basis with over 80 validated human-susceptibility loci [7]. Among them are polymorphisms in the endogenous retrovirus HRES-1 locus. T cells of SLE patients overexpress HRES-1/Rab4, depleting Drp1 levels, which impairs mitophagy, resulting in the accumulation of defective mitochondria [8]. Treatment with a Rab4 inhibitor restored Drp1 expression and decreased autoimmune pathology in lupus-prone mice, validating this pathway in lupus

Mitochondria dominance in lupus metabolism

Multiple studies using a broad variety of approaches have identified defective mitochondria as a hub of metabolic defects in lupus, which has been the topic of recent reviews 58, 59. Here, we review recent studies that have uncovered novel complex, cell-specific cellular and molecular pathways by which mitochondria contribute to lupus pathogenesis (Figure 2).

Future directions

A requirement for large cell numbers has hampered the metabolomic analysis of immune cells, which present an array of developmental and functional subsets, each of which with probably specific metabolic requirements. Approaches developed to conduct single-cell metabolism 81, 82 have started to be applied to immune cells, such as in vitro-polarized human macrophages [83], and they may be used in autoimmunity research in the near future. Technological advances have also produced promising results

Declaration of competing interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgements

This work was supported by the National Institutes of Health RO1AI128901, RO1AI045050, and RO1AI143313.

References and recommended reading (88)

  • K.A. Fortner et al.

    Targeting mitochondrial oxidative stress with MitoQ reduces NET formation and kidney disease in lupus-prone MRL-lpr mice

    Lupus Sci Med

    (2020)
  • D.M. Patrick et al.

    Isolevuglandins disrupt PU.1-mediated C1q expression and promote autoimmunity and hypertension in systemic lupus erythematosus

    JCI Insight

    (2022)
  • E. Katsuyama et al.

    The CD38/NAD/SIRTUIN1/EZH2 axis mitigates cytotoxic CD8 T cell function and identifies patients with SLE prone to infections

    Cell Rep

    (2020)
  • J. Wu et al.

    Boosting NAD+ blunts TLR4-induced type I IFN in control and systemic lupus erythematosus monocytes

    J Clin Investig

    (2022)
  • M. Terrell et al.

    The intersection of cellular and systemic metabolism: Metabolic syndrome in systemic lupus erythematosus

    Endocrinol

    (2022)
  • M. Vukelic et al.

    T cell Metabolism in Lupus

    Immunometabolism

    (2020)
  • X. Teng et al.

    Metabolic determinants of lupus pathogenesis

    Immunol Rev

    (2020)
  • P. Piranavan et al.

    Metabolic targets for treatment of autoimmune diseases

    Immunometabolism

    (2020)
  • L. Chen et al.

    Genetic advances in systemic lupus erythematosus: an update

    Curr Opin Rheuma

    (2017)
  • T.N. Caza et al.

    HRES-1/Rab4-mediated depletion of Drp1 impairs mitochondrial homeostasis and represents a target for treatment in SLE

    Ann Rheum Dis

    (2014)
  • D.J. Perry et al.

    Murine lupus susceptibility locus Sle1c2 mediates CD4+ T cell activation and maps to estrogen-related receptor gamma

    J Immunol

    (2012)
  • W. Li et al.

    Lupus susceptibility gene Esrrg modulates regulatory T cells through mitochondrial metabolism

    JCI Insight

    (2021)
  • R.D. Michalek et al.

    Cutting edge: distinct glycolytic and lipid oxidative metabolic programs are essential for effector and regulatory CD4+ T cell subsets

    J Immunol

    (2011)
  • P. Dhillon et al.

    The nuclear receptor Esrra protects from kidney disease by coupling metabolism and differentiation

    Cell Metab

    (2021)
  • S. Hong et al.

    The emerging role of renal tubular epithelial cells in the immunological pathophysiology of lupus nephritis

    Front Immunol

    (2020)
  • H. Li et al.

    IL-23 reshapes kidney resident cell metabolism and promotes local kidney inflammation

    J Clin Investig

    (2021)
  • C.A. Schnabel et al.

    Expression of Pbx1b during mammalian organogenesis

    Mech Dev

    (2001)
  • C.M. Cuda et al.

    Pre-B cell leukemia homeobox 1 is associated with lupus susceptibility in mice and humans

    J Immunol

    (2012)
  • S.K. Kim et al.

    Pbx1 inactivation disrupts pancreas development and in Ipf1-deficient mice promotes diabetes mellitus

    Nat Genet

    (2002)
  • Y. Jiang et al.

    PBX homeobox 1 enhances hair follicle mesenchymal stem cell proliferation and reprogramming through activation of the AKT/glycogen synthase kinase signaling pathway and suppression of apoptosis

    Stem Cell Res Ther

    (2019)
  • H.Y. Chung et al.

    Meis1, Hi1α, and GATA1 are integrated into a hierarchical regulatory network to mediate primitive erythropoiesis

    FASEB J

    (2021)
  • T. Roach et al.

    Genetic variations controlling regulatory T cell development and activity in mouse models of lupus-like autoimmunity

    Front Immunol

    (2022)
  • T. Katsuyama et al.

    Splicing factor SRSF1 controls T cell hyperactivity and systemic autoimmunity

    J Clin Investig

    (2019)
  • B.I. Reinfeld et al.

    Cell-programmed nutrient partitioning in the tumour microenvironment

    Nature

    (2021)
  • R.D. Leone et al.

    Glutamine blockade induces divergent metabolic programs to overcome tumor immune evasion

    Science

    (2019)
  • S.C. Choi et al.

    Inhibition of glucose metabolism selectively targets autoreactive follicular helper T cells

    Nat Commun

    (2018)
  • X. Zou et al.

    Metabolic regulation of follicular helper T cell differentiation in a mouse model of lupus

    Immunol Lett

    (2022)
  • S.C. Choi et al.

    Pharmacologically inferred glycolysis and glutaminolysis requirement of B cells in lupus-prone mice

    J Immunol

    (2022)
  • F.J. Weisel et al.

    Germinal center B cells selectively oxidize fatty acids for energy while conducting minimal glycolysis

    Nat Immunol

    (2020)
  • C.P. Martins et al.

    Glycolysis inhibition induces functional and metabolic exhaustion of CD4+ T cells in type 1 diabetes

    Front Immunol

    (2021)
  • N.E. Scharping et al.

    Mitochondrial stress induced by continuous stimulation under hypoxia rapidly drives T cell exhaustion

    Nat Immunol

    (2021)
  • D.R. Wahl et al.

    Characterization of the metabolic phenotype of chronically activated lymphocytes

    Lupus

    (2010)
  • C.G. Weindel et al.

    B cell autophagy mediates TLR7-dependent autoimmunity and inflammation

    Autophagy

    (2015)
  • B.M. Miller et al.

    The habitat filters of microbiota-nourishing immunity

    Annu Rev Immunol

    (2021)
  • Cited by (5)

    View full text