You are on page 1of 2

ll

Previews
Research Council of Canada under award number (2020). Severe reactive astrocytes precipitate path- tiomers. Toxicol. Sci. 56, 124–132. https://doi.org/
RGPIN-2019-355803. A.K.F. received an Ontario ological hallmarks of Alzheimer’s disease via 10.1093/toxsci/56.1.124.
Graduate Scholarship and Postgraduate Scholar- H2O2 production. Nat. Neurosci. 23, 1555–1566.
ship – Doctoral from the Natural Sciences and En- https://doi.org/10.1038/s41593-020-00735-y. Pendeville, H., Carpino, N., Marine, J.-C.,
gineering Research Council of Canada. Takahashi, Y., Muller, M., Martial, J.A., and
Jo, S., Yarishkin, O., Hwang, Y.J., Chun, Y.E., Park, Cleveland, J.L. (2001). The ornithine decarboxy-
M., Woo, D.H., Bae, J.Y., Kim, T., Lee, J., Chun, H., lase gene is essential for cell survival during early
DECLARATION OF INTERESTS et al. (2014). GABA from reactive astrocytes im- murine development. Mol. Cell Biol. 21, 6549–
pairs memory in mouse models of Alzheimer’s dis- 6558. https://doi.org/10.1128/MCB.21.19.6549-
The authors declare no competing interests. ease. Nat. Med. 20, 886–896. https://doi.org/10. 6558.2001.
1038/nm.3639.
REFERENCES Ju, Y.H., Bhalla, M., Hyeon, S.J., Oh, J.E., Yoo, S., Price, B.R., Johnson, L.A., and Norris, C.M. (2021).
Chae, U., Kwon, J., Koh, W., Lim, J., Park, Y.M., Reactive astrocytes: the nexus of pathological and
et al. (2022). Astrocytic urea cycle detoxifies clinical hallmarks of Alzheimer’s disease. Ageing
Alber, J., McGarry, K., Noto, R.B., and Snyder, P.J.
Ab-derived ammonia while impairing memory in Res. Rev. 68, 101335. https://doi.org/10.1016/j.
(2018). Use of eflornithine (DFMO) in the treatment
Alzheimer’s disease. Cell Metab. 34, 1104–1120. arr.2021.101335.
of early Alzheimer’s disease: a compassionate use,
single-case study. Front. Aging Neurosci. 10, 60. https://doi.org/10.1016/j.cmet.2022.05.011.
https://doi.org/10.3389/fnagi.2018.00060. Xu, J., Begley, P., Church, S.J., Patassini, S.,
Kim, C.K., Lee, Y.R., Ong, L., Gold, M., Kalali, A., Hollywood, K.A., Ju €llig, M., Curtis, M.A.,
Chun, H., and Lee, C.J. (2018). Reactive astrocytes and Sarkar, J. (2022). Alzheimer’s disease: key in- Waldvogel, H.J., Faull, R.L., Unwin, R.D., and
in Alzheimer’s disease: a double-edged sword. sights from two decades of clinical trial failures. Cooper, G.J. (2016). Graded perturbations of
Neurosci. Res. 126, 44–52. https://doi.org/10. J. Alzheim. Dis. 87, 83–100. https://doi.org/10. metabolism in multiple regions of human brain in
1016/j.neures.2017.11.012. 3233/JAD-215699. Alzheimer’s disease: snapshot of a pervasive
metabolic disorder. Biochim. Biophys. Acta.
Chun, H., Im, H., Kang, Y.J., Kim, Y., Shin, J.H., McWilliams, M.L. (2000). Characterization of the 1862, 1084–1092. https://doi.org/10.1016/j.bba-
Won, W., Lim, J., Ju, Y., Park, Y.M., Kim, S., et al. ototoxicity of difluoromethylornithine and its enan- dis.2016.03.001.

T-bet B or not T-bet B, is that the question


in obesity-related metabolic disease?
Michael P. Cancro1,*
1Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA

*Correspondence: cancro@pennmedicine.upenn.edu
https://doi.org/10.1016/j.cmet.2022.07.008

Obesity is linked to inflammation and downstream metabolic dysregulation. In this issue of Cell Metabolism,
Ha€gglöf et al. show that iNKT cells enable the accumulation of T-bet+ B cells in white adipose tissue, which in
turn produce chemokine and antibody mediators that exacerbate the onset and severity of metabolic
disease.

Although associations between obesity, the T-bet+ B lymphocyte subset, and authors note that CD1d, a non-canonical
inflammatory cytokines, and metabolic innate natural killer T (iNKT) cells. More- antigen-presenting molecule that inter-
disease have been well established, a over, they show that these T-bet+ B cells acts preferentially with iNKT cells,
growing literature suggests that these are causally related to the onset and was downregulated on T-bet+ B cells
processes might also involve interactions progression of metabolic disease in a compared to their T-bet counterparts.
with cells of the innate and adaptive im- mouse model. These core observations prompted them
mune systems. For example, leukocytes The primary findings driving these to ask what drives T-bet+ B cell expansion
or lymphocytes infiltrate many target tis- studies are drawn from patient samples in this setting and what role they play in
sues in these diseases, and antibodies and show that in white adipose tissue the establishment of obesity-associated
to various self-components are frequently from individuals with obesity, the repre- metabolic disease.
observed. Against this backdrop, Ha €gglöf sentation of T-bet+ B cells is elevated T-bet+ B cells have been increasingly
et al. (2022) sought to assess whether B and strongly correlated with body mass scrutinized over the last decade and are
lymphocytes are involved in obesity- index (BMI) and weight. More extensive variously termed age-associated B cells
driven metabolic disease, and to establish analyses of these cells reveal additional (Hao et al., 2011; Rubtsov et al., 2011),
the cellular and molecular interactions surface and signaling molecules associ- double-negative B cells (Jenks et al.,
involved. In toto, their findings reveal a ated with T-bet+ B cells, including the in- 2018), atypical memory B cells (Obeng-
previously unappreciated circuit of inter- tegrin CD11c and several indicators of Adjei et al., 2017), or simply T-bet+ mem-
actions between white adipose tissue, recent or ongoing activation. Finally, the ory B cells (Winslow et al., 2017). They are

Cell Metabolism 34, August 2, 2022 ª 2022 Elsevier Inc. 1081


ll
Previews

associated with both microbe-specific quent production of IFN-g, a known driver phage recruitment suggests this triad
immunity and vaccination but are also of T-bet+ B cell fate. could be involved in other obesity-related
expanded in numerous autoimmune dis- Returning to their core observations, diseases, such as cardiovascular syn-
eases (reviewed in Cancro, 2020). Despite the authors ultimately asked whether dromes and atherosclerosis. These and
increasing appreciation for T-bet+ B cells and how T-bet+ B cells affect the onset related questions provide ample opportu-
in these settings, their potential roles in or severity of metabolic disease in the nity for experimental interrogation going
obesity and metabolic disease have not context of obesity. Using knockout and forward.
been explored in detail. Several groups adoptive transfer strategies, the authors
have previously reported B cells with show that the absence of T-bet+ B cells DECLARATION OF INTERESTS
similar characteristics in human adipose reduces the prevalence and onset of
The author declares no competing interests.
tissue (Frasca et al., 2021), but the obser- metabolic disease, and that disease is
vations reported by Ha €gglöf et al. sub- restored upon the adoptive transfer of pu- REFERENCES
stantially extend earlier descriptions. rified IgG from HFD-fed WT mice. These
To interrogate the nature, interactions, key observations forge a causal link be- Cancro, M.P. (2020). Age-associated B cells.
and roles of T-bet+ B cells more precisely tween obesity-associated metabolic dis- Annu. Rev. Immunol. 38, 315–340. https://doi.
org/10.1146/annurev-immunol-092419-031130.
in obesity-related metabolic disease, the ease and T-bet+ B cells.
authors turned to a murine model wherein In aggregate, these results provide Frasca, D., Romero, M., Diaz, A., Garcia, D.,
Thaller, S., and Blomberg, B.B. (2021). B cells
these features are induced by a high-fat mechanistic insight into the origin and with a senescent-associated secretory phenotype
diet (HFD). Analogous to the findings in role of adipose tissue-associated T-bet+ accumulate in the adipose tissue of individuals with
humans, the proportions and numbers of B cells in metabolic disease. In addition, obesity. Int. J. Mol. Sci. 22, 1839. https://doi.org/
10.3390/ijms22041839.
T-bet+ B cells were expanded in adipose they have clear implications for targeting
tissues and spleens of mice fed an HFD. either the cells themselves or the interac- Ha€gglöf, T., Vanz, C., Kumagai, A., Dudley, E.,
Ortega, V., Siller, M., Parthasarathy, R., Keegan,
This was accompanied by elevations in tions involved in their generation as J., Koenigs, A., Shute, T., and Leadbetter, E.A.
the levels of circulating IgG2c, the potential interventions. These might (2022). T-bet+ B cells accumulate in adipose tissue
murine immunoglobulin heavy chain iso- involve directly targeting T-bet+ cells and exacerbate metabolic disorder during obesity.
Cell Metab. 34, 1121–1136.
type characteristically produced by through unique surface molecules,
T-bet+ B cells. Importantly, the expanded leveraging unique metabolic properties, Hao, Y., O’Neill, P., Naradikian, M.S., Scholz, J.L.,
and Cancro, M.P. (2011). A B-cell subset uniquely
T-bet+ B cells showed phenotypic and or blocking the molecular interactions responsive to innate stimuli accumulates in aged
activation characteristics similar to their required for their generation. Whether mice. Blood 118, 1294–1304. https://doi.org/10.
human counterparts, including the altered such approaches could prevent, arrest, 1182/blood-2011-01-330530.
expression of CD1d. Intrigued by this or reverse disease remains speculative Jenks, S.A., Cashman, K.S., Zumaquero, E.,
observation and given the unique interac- but constitutes potentially fertile transla- Marigorta, U.M., Patel, A.V., Wang, X., Tomar, D.,
Woodruff, M.C., Simon, Z., Bugrovsky, R., et al.
tions of CD1d with iNKT cells, the authors tional ground. (2018). Distinct effector B cells induced by un-
directly asked whether iNKT cells might Several important fundamental ques- regulated Toll-like receptor 7 contribute to patho-
be crucial for generating adipose tissue tions remain. Since IgG2c antibodies per genic responses in systemic lupus erythematosus.
Immunity 49, 725–739.e6. https://doi.org/10.1016/
T-bet+ B cells by using iNKT cell-deficient se are implicated, determining the events j.immuni.2018.08.015.
CD1d1 knockouts. Strikingly, in contrast and signals that drive T-bet+ B cell differ-
Obeng-Adjei, N., Portugal, S., Holla, P., Li, S.,
to HFD-fed wild-type (WT) mice, neither entiation to plasma cells is an important Sohn, H., Ambegaonkar, A., Skinner, J., Bowyer,
the expansion of T-bet+ B cells nor the step in completing the circle. However, G., Doumbo, O.K., Traore, B., et al. (2017).
increase in IgG2c occurred in iNKT cell- because plasma cells do not express Malaria-induced interferon-gamma drives the
expansion of Tbethi atypical memory B cells.
deficient mice. T-bet, studies probing this question will PLoS Pathog. 13, e1006576. https://doi.org/10.
To parse the events involved in these in- likely require genetic tools that afford lon- 1371/journal.ppat.1006576.
teractions, the authors employed in vivo gitudinal fate mapping. In a broader Rubtsov, A.V., Rubtsova, K., Fischer, A., Meehan,
models in which parameters key to the context, these findings raise the question R.T., Gillis, J.Z., Kappler, J.W., and Marrack, P.
generation of T-bet+ B cells could be of whether the T-bet+ B cell subset— (2011). Toll-like receptor 7 (TLR7)-driven accumu-
lation of a novel CD11c(+) B-cell population is
manipulated in the context of iNKT recog- which has already been implicated in important for the development of autoimmunity.
nition of CD1d-presented glycolipid. numerous autoimmune and autoinflam- Blood 118, 1305–1315. https://doi.org/10.1182/
Collectively, these experiments show matory diseases—extends to additional blood-2011-01-331462.

that the in vivo generation and expansion autoinflammatory diseases related to Winslow, G.M., Papillion, A.M., Kenderes, K.J.,
of T-bet+ B cells by TLR7-driven activa- obesity and metabolism. For example, and Levack, R.C. (2017). CD11c+ T-bet+ memory
B cells: immune maintenance during chronic infec-
tion require iNKT cells, likely reflecting the link between T-bet+ B cells, their anti- tion and inflammation? Cell. Immunol. 321, 8–17.
cognate interactions via CD1d and subse- body products, and inflammatory macro- https://doi.org/10.1016/j.cellimm.2017.07.006.

1082 Cell Metabolism 34, August 2, 2022

You might also like