Honors & Awards


  • SNF postdoc fellowship, Schweizerische Nationalfonds (Swiss National Science Foundation) (2023)
  • Pfizer Research Prize, Stiftung Pfizer Forschungspreis (Pfizer Research Prize Foundation) (2022)
  • Science & SciLifeLab Prize for Young Scientists, Science/AAAS, SciLifeLab (2021)
  • SNF postdoc fellowship, Schweizerische Nationalfonds (Swiss National Science Foundation) (2021)
  • ETH Medal for outstanding doctoral theses, ETH Zürich (Swiss Federal Institute of Technology in Zürich) (2020)
  • Award for Outstanding Self-Financed Students Abroad, Ministry of Education of China (2019)
  • Best poster award (1st prize) in 58th International Conference on the Bioscience of Lipids, The International Conference on the Bioscience of Lipids (ICBL) (2017)

Professional Education


  • Doctor of Philosophy, ETH Zurich (2020)
  • Master of Science, Columbia University (2014)
  • Bachelor of Science, China Pharmaceutical University (2010)

Stanford Advisors


Lab Affiliations


All Publications


  • Spatial transcriptomics reveal neuron-astrocyte synergy in long-term memory. Nature Sun, W., Liu, Z., Jiang, X., Chen, M. B., Dong, H., Liu, J., Südhof, T. C., Quake, S. R. 2024

    Abstract

    Memory encodes past experiences, thereby enabling future plans. The basolateral amygdala is a centre of salience networks that underlie emotional experiences and thus has a key role in long-term fear memory formation1. Here we used spatial and single-cell transcriptomics to illuminate the cellular and molecular architecture of the role of the basolateral amygdala in long-term memory. We identified transcriptional signatures in subpopulations of neurons and astrocytes that were memory-specific and persisted for weeks. These transcriptional signatures implicate neuropeptide and BDNF signalling, MAPK and CREB activation, ubiquitination pathways, and synaptic connectivity as key components of long-term memory. Notably, upon long-term memory formation, a neuronal subpopulation defined by increased Penk and decreased Tac expression constituted the most prominent component of the memory engram of the basolateral amygdala. These transcriptional changes were observed both with single-cell RNA sequencing and with single-molecule spatial transcriptomics in intact slices, thereby providing a rich spatial map of a memory engram. The spatial data enabled us to determine that this neuronal subpopulation interacts with adjacent astrocytes, and functional experiments show that neurons require interactions with astrocytes to encode long-term memory.

    View details for DOI 10.1038/s41586-023-07011-6

    View details for PubMedID 38326616

    View details for PubMedCentralID 4292167

  • Identification of a regulatory pathway inhibiting adipogenesis via RSPO2. Nature metabolism Dong, H., Sun, W., Shen, Y., Baláz, M., Balázová, L., Ding, L., Löffler, M., Hamilton, B., Klöting, N., Blüher, M., Neubauer, H., Klein, H., Wolfrum, C. 2022

    Abstract

    Healthy adipose tissue remodeling depends on the balance between de novo adipogenesis from adipogenic progenitor cells and the hypertrophy of adipocytes. De novo adipogenesis has been shown to promote healthy adipose tissue expansion, which confers protection from obesity-associated insulin resistance. Here, we define the role and trajectory of different adipogenic precursor subpopulations and further delineate the mechanism and cellular trajectory of adipogenesis, using single-cell RNA-sequencing datasets of murine adipogenic precursors. We identify Rspo2 as a functional regulator of adipogenesis, which is secreted by a subset of CD142+ cells to inhibit maturation of early progenitors through the receptor Lgr4. Increased circulating RSPO2 in mice leads to adipose tissue hypertrophy and insulin resistance and increased RSPO2 levels in male obese individuals correlate with impaired glucose homeostasis. Taken together, these findings identify a complex cellular crosstalk that inhibits adipogenesis and impairs adipose tissue homeostasis.

    View details for DOI 10.1038/s42255-021-00509-1

    View details for PubMedID 35027768

  • Analysis of Single-Cell/Nucleus Transcriptome Data in Adipose Tissue. Methods in molecular biology (Clifton, N.J.) Sun, W. 2022; 2448: 291-306

    Abstract

    Adipose tissue is highly heterogeneous and plastic. Recent advances in single-cell/nucleus RNA sequencing technology have helped to study the cellular composition and dynamics of adipose tissue. In this protocol, I outline a typical workflow of analyzing single-cell/nucleus transcriptome data. Specifically, I show an example of how cellular populations are estimated and characterized from a single-nucleus RNAseq data set of frozen archived human adipose tissue.

    View details for DOI 10.1007/978-1-0716-2087-8_19

    View details for PubMedID 35167105

  • Local acetate inhibits brown adipose tissue function. Proceedings of the National Academy of Sciences of the United States of America Sun, W., Dong, H., Wolfrum, C. 2021; 118 (49)

    Abstract

    Brown adipose tissue has been extensively studied in the last decade for its potential to counteract the obesity pandemic. However, the paracrine regulation within brown tissue is largely unknown. Here, we show that local acetate directly inhibits brown fat thermogenesis, without changing acetate levels in the circulation. We demonstrate that modulating acetate within brown tissue at physiological levels blunts its function and systemically decreases energy expenditure. Using a series of transcriptomic analyses, we identified genes related to the tricarboxylic acid cycle and brown adipocyte formation, which are down-regulated upon local acetate administration. Overall, these findings demonstrate that local acetate inhibits brown fat function.

    View details for DOI 10.1073/pnas.2116125118

    View details for PubMedID 34845033

  • Fat for heat. Science (New York, N.Y.) Sun, W. 2021; 374 (6571): 1066

    Abstract

    [Figure: see text].

    View details for DOI 10.1126/science.abm8138

    View details for PubMedID 34822283

  • Plasticity and heterogeneity of thermogenic adipose tissue NATURE METABOLISM Sun, W., Modica, S., Dong, H., Wolfrum, C. 2021; 3 (6): 751-761

    Abstract

    The perception of adipose tissue, both in the scientific community and in the general population, has changed dramatically in the past 20 years. While adipose tissue was thought for a long time to be a rather simple lipid storage entity, it is now recognized as a highly heterogeneous organ and a critical regulator of systemic metabolism, composed of many different subtypes of cells, with important endocrine functions. Additionally, adipose tissue is nowadays recognized to contribute to energy turnover, due to the presence of specialized thermogenic adipocytes, which can be found in many adipose depots. This review discusses the unprecedented insights that we have gained into the heterogeneity of thermogenic adipocytes and their respective precursors due to the technical developments in single-cell and nucleus technologies. These methodological advances have increased our understanding of how adipose tissue catabolic function is influenced by developmental and intercellular communication events.

    View details for DOI 10.1038/s42255-021-00417-4

    View details for Web of Science ID 000665078900005

    View details for PubMedID 34158657

  • snRNA-seq reveals a subpopulation of adipocytes that regulates thermogenesis NATURE Sun, W., Dong, H., Balaz, M., Slyper, M., Drokhlyansky, E., Colleluori, G., Giordano, A., Kovanicova, Z., Stefanicka, P., Balazova, L., Ding, L., Husted, A., Rudofsky, G., Ukropec, J., Cinti, S., Schwartz, T. W., Regev, A., Wolfrum, C. 2020; 587 (7832): 98-+

    Abstract

    Adipose tissue is usually classified on the basis of its function as white, brown or beige (brite)1. It is an important regulator of systemic metabolism, as shown by the fact that dysfunctional adipose tissue in obesity leads to a variety of secondary metabolic complications2,3. In addition, adipose tissue functions as a signalling hub that regulates systemic metabolism through paracrine and endocrine signals4. Here we use single-nucleus RNA-sequencing (snRNA-seq) analysis in mice and humans to characterize adipocyte heterogeneity. We identify a rare subpopulation of adipocytes in mice that increases in abundance at higher temperatures, and we show that this subpopulation regulates the activity of neighbouring adipocytes through acetate-mediated modulation of their thermogenic capacity. Human adipose tissue contains higher numbers of cells of this subpopulation, which could explain the lower thermogenic activity of human compared to mouse adipose tissue and suggests that targeting this pathway could be used to restore thermogenic activity.

    View details for DOI 10.1038/s41586-020-2856-x

    View details for Web of Science ID 000582810000003

    View details for PubMedID 33116305

  • Environmental and Nutritional Effects Regulating Adipose Tissue Function and Metabolism Across Generations ADVANCED SCIENCE Sun, W., von Meyenn, F., Peleg-Raibstein, D., Wolfrum, C. 2019; 6 (11): 1900275

    Abstract

    The unabated rise in obesity prevalence during the last 40 years has spurred substantial interest in understanding the reasons for this epidemic. Studies in mice and humans have demonstrated that obesity is a highly heritable disease; however genetic variations within specific populations have so far not been able to explain this phenomenon to its full extent. Recent work has demonstrated that environmental cues can be sensed by an organism to elicit lasting changes, which in turn can affect systemic energy metabolism by different epigenetic mechanisms such as changes in small noncoding RNA expression, DNA methylation patterns, as well as histone modifications. These changes can directly modulate cellular function in response to environmental cues, however research during the last decade has demonstrated that some of these modifications might be transmitted to subsequent generations, thus modulating energy metabolism of the progeny in an inter- as well as transgenerational manner. In this context, adipose tissue has become a focus of research due to its plasticity, which allows the formation of energy storing (white) as well as energy wasting (brown/brite/beige) cells within the same depot. In this Review, the effects of environmental induced obesity with a particular focus on adipose tissue are discussed.

    View details for DOI 10.1002/advs.201900275

    View details for Web of Science ID 000470189500025

    View details for PubMedID 31179229

    View details for PubMedCentralID PMC6548959

  • Fat cells with a sweet tooth NATURE Sun, W., Wolfrum, C. 2019; 565 (7738): 167-168

    View details for DOI 10.1038/d41586-018-07739-6

    View details for Web of Science ID 000455231000031

    View details for PubMedID 30622347

  • Cold-induced epigenetic programming of the sperm enhances brown adipose tissue activity in the offspring NATURE MEDICINE Sun, W., Dong, H., Becker, A. S., Dapito, D. H., Modica, S., Grandl, G., Opitz, L., Efthymiou, V., Straub, L. G., Sarker, G., Balaz, M., Balazova, L., Perdikari, A., Kiehlmann, E., Bacanovic, S., Zellweger, C., Peleg-Raibstein, D., Pelczar, P., Reik, W., Burger, I. A., von Meyenn, F., Wolfrum, C. 2018; 24 (9): 1372-+

    Abstract

    Recent research has focused on environmental effects that control tissue functionality and systemic metabolism. However, whether such stimuli affect human thermogenesis and body mass index (BMI) has not been explored. Here we show retrospectively that the presence of brown adipose tissue (BAT) and the season of conception are linked to BMI in humans. In mice, we demonstrate that cold exposure (CE) of males, but not females, before mating results in improved systemic metabolism and protection from diet-induced obesity of the male offspring. Integrated analyses of the DNA methylome and RNA sequencing of the sperm from male mice revealed several clusters of co-regulated differentially methylated regions (DMRs) and differentially expressed genes (DEGs), suggesting that the improved metabolic health of the offspring was due to enhanced BAT formation and increased neurogenesis. The conclusions are supported by cell-autonomous studies in the offspring that demonstrate an enhanced capacity to form mature active brown adipocytes, improved neuronal density and more norepinephrine release in BAT in response to cold stimulation. Taken together, our results indicate that in humans and in mice, seasonal or experimental CE induces an epigenetic programming of the sperm such that the offspring harbor hyperactive BAT and an improved adaptation to overnutrition and hypothermia.

    View details for DOI 10.1038/s41591-018-0102-y

    View details for Web of Science ID 000444174100018

    View details for PubMedID 29988127

  • Adipocyte p53 coordinates the response to intermittent fasting by regulating adipose tissue immune cell landscape. Nature communications Reinisch, I., Michenthaler, H., Sulaj, A., Moyschewitz, E., Krstic, J., Galhuber, M., Xu, R., Riahi, Z., Wang, T., Vujic, N., Amor, M., Zenezini Chiozzi, R., Wabitsch, M., Kolb, D., Georgiadi, A., Glawitsch, L., Heitzer, E., Schulz, T. J., Schupp, M., Sun, W., Dong, H., Ghosh, A., Hoffmann, A., Kratky, D., Hinte, L. C., von Meyenn, F., Heck, A. J., Blüher, M., Herzig, S., Wolfrum, C., Prokesch, A. 2024; 15 (1): 1391

    Abstract

    In obesity, sustained adipose tissue (AT) inflammation constitutes a cellular memory that limits the effectiveness of weight loss interventions. Yet, the impact of fasting regimens on the regulation of AT immune infiltration is still elusive. Here we show that intermittent fasting (IF) exacerbates the lipid-associated macrophage (LAM) inflammatory phenotype of visceral AT in obese mice. Importantly, this increase in LAM abundance is strongly p53 dependent and partly mediated by p53-driven adipocyte apoptosis. Adipocyte-specific deletion of p53 prevents LAM accumulation during IF, increases the catabolic state of adipocytes, and enhances systemic metabolic flexibility and insulin sensitivity. Finally, in cohorts of obese/diabetic patients, we describe a p53 polymorphism that links to efficacy of a fasting-mimicking diet and that the expression of p53 and TREM2 in AT negatively correlates with maintaining weight loss after bariatric surgery. Overall, our results demonstrate that p53 signalling in adipocytes dictates LAM accumulation in AT under IF and modulates fasting effectiveness in mice and humans.

    View details for DOI 10.1038/s41467-024-45724-y

    View details for PubMedID 38360943

    View details for PubMedCentralID PMC10869344

  • Alternative splicing of latrophilin-3 controls synapse formation. Nature Wang, S., DeLeon, C., Sun, W., Quake, S. R., Roth, B. L., Südhof, T. C. 2024

    Abstract

    The assembly and specification of synapses in the brain is incompletely understood1-3. Latrophilin-3 (encoded by Adgrl3, also known as Lphn3)-a postsynaptic adhesion G-protein-coupled receptor-mediates synapse formation in the hippocampus4 but the mechanisms involved remain unclear. Here we show in mice that LPHN3 organizes synapses through a convergent dual-pathway mechanism: activation of Gαs signalling and recruitment of phase-separated postsynaptic protein scaffolds. We found that cell-type-specific alternative splicing of Lphn3 controls the LPHN3 G-protein-coupling mode, resulting in LPHN3 variants that predominantly signal through Gαs or Gα12/13. CRISPR-mediated manipulation of Lphn3 alternative splicing that shifts LPHN3 from a Gαs- to a Gα12/13-coupled mode impaired synaptic connectivity as severely as the overall deletion of Lphn3, suggesting that Gαs signalling by LPHN3 splice variants mediates synapse formation. Notably, Gαs-coupled, but not Gα12/13-coupled, splice variants of LPHN3 also recruit phase-transitioned postsynaptic protein scaffold condensates, such that these condensates are clustered by binding of presynaptic teneurin and FLRT ligands to LPHN3. Moreover, neuronal activity promotes alternative splicing of the synaptogenic Gαs-coupled variant of LPHN3. Together, these data suggest that activity-dependent alternative splicing of a key synaptic adhesion molecule controls synapse formation by parallel activation of two convergent pathways: Gαs signalling and clustered phase separation of postsynaptic protein scaffolds.

    View details for DOI 10.1038/s41586-023-06913-9

    View details for PubMedID 38233523

    View details for PubMedCentralID 8186004

  • Laminin α4 Expression in Human Adipose Tissue Depots and Its Association with Obesity and Obesity Related Traits. Biomedicines Hagemann, T., Czechowski, P., Ghosh, A., Sun, W., Dong, H., Noé, F., Wolfrum, C., Blüher, M., Hoffmann, A. 2023; 11 (10)

    Abstract

    Laminin α4 (LAMA4) is one of the main structural adipocyte basement membrane (BM) components that is upregulated during adipogenesis and related to obesity in mice and humans. We conducted RNA-seq-based gene expression analysis of LAMA4 in abdominal subcutaneous (SC) and visceral (VIS) adipose tissue (AT) depots across three human sub-cohorts of the Leipzig Obesity BioBank (LOBB) to explore the relationship between LAMA4 expression and obesity (N = 1479) in the context of weight loss (N = 65) and metabolic health (N = 42). We found significant associations of LAMA4 with body fat mass (p < 0.001) in VIS AT; higher expression in VIS AT compared to SC AT; and significant relation to metabolic health parameters e.g., body fat in VIS AT, waist (p = 0.009) and interleukin 6 (p = 0.002) in male VIS AT, and hemoglobin A1c (p = 0.008) in male SC AT. AT LAMA4 expression was not significantly different between subjects with or without obesity, metabolically healthy versus unhealthy, and obesity before versus after short-term weight loss. Our results support significant associations between obesity related clinical parameters and elevated LAMA4 expression in humans. Our work offers one of the first references for understanding the meaning of LAMA4 expression specifically in relation to obesity based on large-scale RNA-seq data.

    View details for DOI 10.3390/biomedicines11102806

    View details for PubMedID 37893179

    View details for PubMedCentralID PMC10604865

  • The obesity-linked human lncRNA AATBC stimulates mitochondrial function in adipocytes. EMBO reports Giroud, M., Kotschi, S., Kwon, Y., Le Thuc, O., Hoffmann, A., Gil-Lozano, M., Karbiener, M., Higareda-Almaraz, J. C., Khani, S., Tews, D., Fischer-Posovszky, P., Sun, W., Dong, H., Ghosh, A., Wolfrum, C., Wabitsch, M., Virtanen, K. A., Blüher, M., Nielsen, S., Zeigerer, A., García-Cáceres, C., Scheideler, M., Herzig, S., Bartelt, A. 2023: e57600

    Abstract

    Adipocytes are critical regulators of metabolism and energy balance. While white adipocyte dysfunction is a hallmark of obesity-associated disorders, thermogenic adipocytes are linked to cardiometabolic health. As adipocytes dynamically adapt to environmental cues by functionally switching between white and thermogenic phenotypes, a molecular understanding of this plasticity could help improving metabolism. Here, we show that the lncRNA Apoptosis associated transcript in bladder cancer (AATBC) is a human-specific regulator of adipocyte plasticity. Comparing transcriptional profiles of human adipose tissues and cultured adipocytes we discovered that AATBC was enriched in thermogenic conditions. Using primary and immortalized human adipocytes we found that AATBC enhanced the thermogenic phenotype, which was linked to increased respiration and a more fragmented mitochondrial network. Expression of AATBC in adipose tissue of mice led to lower plasma leptin levels. Interestingly, this association was also present in human subjects, as AATBC in adipose tissue was inversely correlated with plasma leptin levels, BMI, and other measures of metabolic health. In conclusion, AATBC is a novel obesity-linked regulator of adipocyte plasticity and mitochondrial function in humans.

    View details for DOI 10.15252/embr.202357600

    View details for PubMedID 37671834

  • Effect of high-dose glucocorticoid treatment on human brown adipose tissue activity: a randomised, double-blinded, placebo-controlled cross-over trial in healthy men. EBioMedicine Maushart, C. I., Sun, W., Othman, A., Ghosh, A., Senn, J. R., Fischer, J. G., Madoerin, P., Loeliger, R. C., Benz, R. M., Takes, M., Zech, C. J., Chirindel, A., Beuschlein, F., Reincke, M., Wild, D., Bieri, O., Zamboni, N., Wolfrum, C., Betz, M. J. 2023; 96: 104771

    Abstract

    BACKGROUND: Glucocorticoids (GCs) are widely applied anti-inflammatory drugs that are associated with adverse metabolic effects including insulin resistance and weight gain. Previous research indicates that GCs may negatively impact brown adipose tissue (BAT) activity in rodents and humans.METHODS: We performed a randomised, double-blinded cross-over trial in 16 healthy men (clinicaltrials.govNCT03269747). Participants received 40mg of prednisone per day for one week or placebo. After a washout period of four weeks, participants crossed-over to the other treatment arm. Primary endpoint was the increase in resting energy expenditure (EE) in response to a mild-cold stimulus (cold-induced thermogenesis, CIT). Secondary outcomes comprised mean 18F-FDG uptake into supraclavicular BAT (SUVmean) as determined by FDG-PET/CT, volume of the BAT depot as well as fat content determined by MRI. The plasma metabolome and the transcriptome of supraclavicular BAT and of skeletal muscle biopsies after each treatment period were analysed.FINDINGS: Sixteen participants were recruited to the trial and completed it successfully per protocol. After prednisone treatment resting EE was higher both during warm and cold conditions. However, CIT was similar, 153 kcal/24h (95% CI 40-266 kcal/24h) after placebo and 186 kcal/24h (95% CI 94-277 kcal/24h, p=0.38) after prednisone. SUVmean of BAT after cold exposure was not significantly affected by prednisone (3.36g/ml, 95% CI 2.69-4.02g/ml, vs 3.07g/ml, 95% CI 2.52-3.62g/ml, p=0.28). Results of plasma metabolomics and BAT transcriptomics corroborated these findings. RNA sequencing of muscle biopsies revealed higher expression of genes involved in calcium cycling. No serious adverse events were reported and adverse events were evenly distributed between the two treatments.INTERPRETATION: Prednisone increased EE in healthy men possibly by altering skeletal muscle calcium cycling. Cold-induced BAT activity was not affected by GC treatment, which indicates that the unfavourable metabolic effects of GCs are independent from thermogenic adipocytes.FUNDING: Grants from Swiss National Science Foundation (PZ00P3_167823), Bangerter-Rhyner Foundation and from Nora van der Meeuwen-Hafliger Foundation to MJB. A fellowship-grant from the Swiss National Science Foundation (SNF211053) to WS. Grants from German Research Foundation (project number: 314061271-TRR 205) and Else Kroner-Fresenius (grant support 2012_A103 and 2015_A228) to MR.

    View details for DOI 10.1016/j.ebiom.2023.104771

    View details for PubMedID 37659283

  • Inhibition of AXL receptor tyrosine kinase enhances brown adipose tissue functionality in mice. Nature communications Efthymiou, V., Ding, L., Balaz, M., Sun, W., Balazova, L., Straub, L. G., Dong, H., Simon, E., Ghosh, A., Perdikari, A., Keller, S., Ghoshdastider, U., Horvath, C., Moser, C., Hamilton, B., Neubauer, H., Wolfrum, C. 2023; 14 (1): 4162

    Abstract

    The current obesity epidemic and high prevalence of metabolic diseases necessitate efficacious and safe treatments. Brown adipose tissue in this context is a promising target with the potential to increase energy expenditure, however no pharmacological treatments activating brown adipose tissue are currently available. Here, we identify AXL receptor tyrosine kinase as a regulator of adipose function. Pharmacological and genetic inhibition of AXL enhance thermogenic capacity of brown and white adipocytes, in vitro and in vivo. Mechanistically, these effects are mediated through inhibition of PI3K/AKT/PDE signaling pathway, resulting in induction of nuclear FOXO1 localization and increased intracellular cAMP levels via PDE3/4 inhibition and subsequent stimulation of the PKA-ATF2 pathway. In line with this, both constitutive Axl deletion as well as inducible adipocyte-specific Axl deletion protect animals from diet-induced obesity concomitant with increases in energy expenditure. Based on these data, we propose AXL receptor as a target for the treatment of obesity.

    View details for DOI 10.1038/s41467-023-39715-8

    View details for PubMedID 37443109

    View details for PubMedCentralID 6099440

  • Adipogenic and SWAT cells separate from a common progenitor in human brown and white adipose depots. Nature metabolism Palani, N. P., Horvath, C., Timshel, P. N., Folkertsma, P., Grønning, A. G., Henriksen, T. I., Peijs, L., Jensen, V. H., Sun, W., Jespersen, N. Z., Wolfrum, C., Pers, T. H., Nielsen, S., Scheele, C. 2023

    Abstract

    Adipocyte function is a major determinant of metabolic disease, warranting investigations of regulating mechanisms. We show at single-cell resolution that progenitor cells from four human brown and white adipose depots separate into two main cell fates, an adipogenic and a structural branch, developing from a common progenitor. The adipogenic gene signature contains mitochondrial activity genes, and associates with genome-wide association study traits for fat distribution. Based on an extracellular matrix and developmental gene signature, we name the structural branch of cells structural Wnt-regulated adipose tissue-resident (SWAT) cells. When stripped from adipogenic cells, SWAT cells display a multipotent phenotype by reverting towards progenitor state or differentiating into new adipogenic cells, dependent on media. Label transfer algorithms recapitulate the cell types in human adipose tissue datasets. In conclusion, we provide a differentiation map of human adipocytes and define the multipotent SWAT cell, providing a new perspective on adipose tissue regulation.

    View details for DOI 10.1038/s42255-023-00820-z

    View details for PubMedID 37337126

    View details for PubMedCentralID 8461455

  • Monoallelic intragenic POU3F2 variants lead to neurodevelopmental delay and hyperphagic obesity, confirming the gene's candidacy in 6q16.1 deletions. American journal of human genetics Schönauer, R., Jin, W., Findeisen, C., Valenzuela, I., Devlin, L. A., Murrell, J., Bedoukian, E. C., Pöschla, L., Hantmann, E., Riedhammer, K. M., Hoefele, J., Platzer, K., Biemann, R., Campeau, P. M., Münch, J., Heyne, H., Hoffmann, A., Ghosh, A., Sun, W., Dong, H., Noé, F., Wolfrum, C., Woods, E., Parker, M. J., Neatu, R., Le Guyader, G., Bruel, A. L., Perrin, L., Spiewak, H., Missotte, I., Fourgeaud, M., Michaud, V., Lacombe, D., Paolucci, S. A., Buchan, J. G., Glissmeyer, M., Popp, B., Blüher, M., Sayer, J. A., Halbritter, J. 2023

    Abstract

    While common obesity accounts for an increasing global health burden, its monogenic forms have taught us underlying mechanisms via more than 20 single-gene disorders. Among these, the most common mechanism is central nervous system dysregulation of food intake and satiety, often accompanied by neurodevelopmental delay (NDD) and autism spectrum disorder. In a family with syndromic obesity, we identified a monoallelic truncating variant in POU3F2 (alias BRN2) encoding a neural transcription factor, which has previously been suggested as a driver of obesity and NDD in individuals with the 6q16.1 deletion. In an international collaboration, we identified ultra-rare truncating and missense variants in another ten individuals sharing autism spectrum disorder, NDD, and adolescent-onset obesity. Affected individuals presented with low-to-normal birth weight and infantile feeding difficulties but developed insulin resistance and hyperphagia during childhood. Except for a variant leading to early truncation of the protein, identified variants showed adequate nuclear translocation but overall disturbed DNA-binding ability and promotor activation. In a cohort with common non-syndromic obesity, we independently observed a negative correlation of POU3F2 gene expression with BMI, suggesting a role beyond monogenic obesity. In summary, we propose deleterious intragenic variants of POU3F2 to cause transcriptional dysregulation associated with hyperphagic obesity of adolescent onset with variable NDD.

    View details for DOI 10.1016/j.ajhg.2023.04.010

    View details for PubMedID 37207645

  • Obesity Is Associated with Distorted Proteoglycan Expression in Adipose Tissue. International journal of molecular sciences Meen, A. J., Doncheva, A. I., Böttcher, Y., Dankel, S. N., Hoffmann, A., Blüher, M., Fernø, J., Mellgren, G., Ghosh, A., Sun, W., Dong, H., Noé, F., Wolfrum, C., Pejler, G., Dalen, K. T., Kolset, S. O. 2023; 24 (8)

    Abstract

    Proteoglycans are central components of the extracellular matrix (ECM) and binding partners for inflammatory chemokines. Morphological differences in the ECM and increased inflammation are prominent features of the white adipose tissues in patients with obesity. The impact of obesity and weight loss on the expression of specific proteoglycans in adipose tissue is not well known. This study aimed to investigate the relationship between adiposity and proteoglycan expression. We analyzed transcriptomic data from two human bariatric surgery cohorts. In addition, RT-qPCR was performed on adipose tissues from female and male mice fed a high-fat diet. Both visceral and subcutaneous adipose tissue depots were analyzed. Adipose mRNA expression of specific proteoglycans, proteoglycan biosynthetic enzymes, proteoglycan partner molecules, and other ECM-related proteins were altered in both human cohorts. We consistently observed more profound alterations in gene expression of ECM targets in the visceral adipose tissues after surgery (among others VCAN (p = 0.000309), OGN (p = 0.000976), GPC4 (p = 0.00525), COL1A1 (p = 0.00221)). Further, gene analyses in mice revealed sex differences in these two tissue compartments in obese mice. We suggest that adipose tissue repair is still in progress long after surgery, which may reflect challenges in remodeling increased adipose tissues. This study can provide the basis for more mechanistic studies on the role of proteoglycans in adipose tissues in obesity.

    View details for DOI 10.3390/ijms24086884

    View details for PubMedID 37108048

    View details for PubMedCentralID PMC10138342

  • A low-carbohydrate diet induces hepatic insulin resistance and metabolic associated fatty liver disease in mice. Molecular metabolism Long, F., Bhatti, M. R., Kellenberger, A., Sun, W., Modica, S., Höring, M., Liebisch, G., Krieger, J. P., Wolfrum, C., Challa, T. D. 2023: 101675

    Abstract

    Metabolic-associated fatty liver disease (MAFLD) is the most common chronic liver disease that can range from hepatic steatosis to non-alcoholic steatohepatitis (NASH), which can lead to fibrosis and cirrhosis. Recently, ketogenic diet (KD), a low carbohydrate diet, gained popularity as a weight-loss approach, although it has been reported to induce hepatic insulin resistance and steatosis in animal model systems via an undefined mechanism. Herein, we investigated the KD metabolic benefits and its contribution to the pathogenesis of NASH.Using metabolic, biochemical and omics approaches, we identified the effects of a KD on NASH and investigated the mechanisms by which KD induces hepatic insulin resistance and steatosis.We demonstrate that KD can induce fibrosis and NASH regardless of body weight loss compared to high-fat diet (HFD) fed mice at thermoneutrality. At ambient temperature (23 °C), KD-fed mice develop a severe hepatic injury, inflammation, and steatosis. In addition, KD increases liver cholesterol, IL-6, and p-JNK and aggravates diet induced-glucose intolerance and hepatic insulin resistance compared to HFD. Pharmacological inhibition of IL-6 and JNK reverses KD-induced glucose intolerance, and hepatic steatosis and restores insulin sensitivity.Our studies uncover a new mechanism for KD-induced hepatic insulin resistance and NASH potentially via IL-6-JNK signaling and provide a new NASH mouse model.

    View details for DOI 10.1016/j.molmet.2023.101675

    View details for PubMedID 36682412

  • Genetic variants in genes involved in creatine biosynthesis in patients with severe obesity or anorexia nervosa. Frontiers in genetics Rajcsanyi, L. S., Hoffmann, A., Ghosh, A., Matrisch-Dinkler, B., Zheng, Y., Peters, T., Sun, W., Dong, H., Noe, F., Wolfrum, C., Herpertz-Dahlmann, B., Seitz, J., de Zwaan, M., Herzog, W., Ehrlich, S., Zipfel, S., Giel, K., Egberts, K., Burghardt, R., Focker, M., Tsai, L. T., Muller, T. D., Bluher, M., Hebebrand, J., Hirtz, R., Hinney, A. 2023; 14: 1128133

    Abstract

    Increased thermogenesis in brown adipose tissue might have an obesity-reducing effect in humans. In transgenic mice, depletion of genes involved in creatine metabolism results in disrupted thermogenic capacity and altered effects of high-fat feeding on body weight. Data analyses of a sex-stratified genome-wide association study (GWAS) for body mass index (BMI) within the genomic regions of genes of this pathway (CKB, CKMT1B, and GATM) revealed one sex-dimorphic BMI-associated SNP in CKB (rs1136165). The effect size was larger in females than in males. A mutation screen of the coding regions of these three candidate genes in a screening group (192 children and adolescents with severe obesity, 192 female patients with anorexia nervosa, and 192 healthy-lean controls) identified five variants in each, CKB and GATM, and nine variants in the coding sequence of CKMT1B. Non-synonymous variants identified in CKB and CKMT1B were genotyped in an independent confirmation study group (781 families with severe obesity (trios), 320 children and adolescents with severe obesity, and 253 healthy-lean controls). In silico tools predicted mainly benign yet protein-destabilizing potentials. A transmission disequilibrium test in trios with severe obesity indicated an obesity-protective effect of the infrequent allele at rs149544188 located in CKMT1B. Subsequent correlation analyses in 1,479 individuals of the Leipzig Obesity BioBank revealed distinct correlations of CKB with the other two genes in omental visceral adipose tissue (VAT) and abdominal subcutaneous adipose tissue (SAT). Furthermore, between-subject comparisons of gene expression levels showed generally higher expressions of all three genes of interest in VAT than in SAT. Future in vitro analyses are needed to assess the functional implications of these findings.

    View details for DOI 10.3389/fgene.2023.1128133

    View details for PubMedID 37101650

  • Single-cell RNA sequencing identifies MFSD2B in megakaryocyte progenitors as a regulator of vascular senescence Atherosclerosis Saeedi, S., Sun, W., Wang, T., Dong, H., Allemann, M., Lee, P., Wolfrum, C., Beer, J. 2023; 379
  • Spatial and single-cell transcriptomics reveal neuron-astrocyte interplay in long-term memory bioRxiv Sun, W., Liu, Z., Jiang, X., Chen, M. B., Dong, H., Liu, J., Südhof, T., Quake, S. 2023
  • Myoglobin-mediated lipid shuttling increases adrenergic activation of brown and white adipocyte metabolism and is as a marker of thermogenic adipocytes in humans. Clinical and translational medicine Christen, L., Broghammer, H., Rapöhn, I., Möhlis, K., Strehlau, C., Ribas-Latre, A., Gebhardt, C., Roth, L., Krause, K., Landgraf, K., Körner, A., Rohde-Zimmermann, K., Hoffmann, A., Klöting, N., Ghosh, A., Sun, W., Dong, H., Wolfrum, C., Rassaf, T., Hendgen-Cotta, U. B., Stumvoll, M., Blüher, M., Heiker, J. T., Weiner, J. 2022; 12 (12): e1108

    Abstract

    Recruitment and activation of brown adipose tissue (BAT) results in increased energy expenditure (EE) via thermogenesis and represents an intriguing therapeutic approach to combat obesity and treat associated diseases. Thermogenesis requires an increased and efficient supply of energy substrates and oxygen to the BAT. The hemoprotein myoglobin (MB) is primarily expressed in heart and skeletal muscle fibres, where it facilitates oxygen storage and flux to the mitochondria during exercise. In the last years, further contributions of MB have been assigned to the scavenging of reactive oxygen species (ROS), the regulation of cellular nitric oxide (NO) levels and also lipid binding. There is a substantial expression of MB in BAT, which is induced during brown adipocyte differentiation and BAT activation. This suggests MB as a previously unrecognized player in BAT contributing to thermogenesis.This study analyzed the consequences of MB expression in BAT on mitochondrial function and thermogenesis in vitro and in vivo. Using MB overexpressing, knockdown or knockout adipocytes, we show that expression levels of MB control brown adipocyte mitochondrial respiratory capacity and acute response to adrenergic stimulation, signalling and lipolysis. Overexpression in white adipocytes also increases their metabolic activity. Mutation of lipid interacting residues in MB abolished these beneficial effects of MB. In vivo, whole-body MB knockout resulted in impaired thermoregulation and cold- as well as drug-induced BAT activation in mice. In humans, MB is differentially expressed in subcutaneous (SC) and visceral (VIS) adipose tissue (AT) depots, differentially regulated by the state of obesity and higher expressed in AT samples that exhibit higher thermogenic potential.These data demonstrate for the first time a functional relevance of MBs lipid binding properties and establish MB as an important regulatory element of thermogenic capacity in brown and likely beige adipocytes.

    View details for DOI 10.1002/ctm2.1108

    View details for PubMedID 36480426

  • Architecture of the outbred brown fat proteome defines regulators of metabolic physiology. Cell Xiao, H., Bozi, L. H., Sun, Y., Riley, C. L., Philip, V. M., Chen, M., Li, J., Zhang, T., Mills, E. L., Emont, M. P., Sun, W., Reddy, A., Garrity, R., Long, J., Becher, T., Vitas, L. P., Laznik-Bogoslavski, D., Ordonez, M., Liu, X., Chen, X., Wang, Y., Liu, W., Tran, N., Liu, Y., Zhang, Y., Cypess, A. M., White, A. P., He, Y., Deng, R., Schöder, H., Paulo, J. A., Jedrychowski, M. P., Banks, A. S., Tseng, Y. H., Cohen, P., Tsai, L. T., Rosen, E. D., Klein, S., Chondronikola, M., McAllister, F. E., Van Bruggen, N., Huttlin, E. L., Spiegelman, B. M., Churchill, G. A., Gygi, S. P., Chouchani, E. T. 2022

    Abstract

    Brown adipose tissue (BAT) regulates metabolic physiology. However, nearly all mechanistic studies of BAT protein function occur in a single inbred mouse strain, which has limited the understanding of generalizable mechanisms of BAT regulation over physiology. Here, we perform deep quantitative proteomics of BAT across a cohort of 163 genetically defined diversity outbred mice, a model that parallels the genetic and phenotypic variation found in humans. We leverage this diversity to define the functional architecture of the outbred BAT proteome, comprising 10,479 proteins. We assign co-operative functions to 2,578 proteins, enabling systematic discovery of regulators of BAT. We also identify 638 proteins that correlate with protection from, or sensitivity to, at least one parameter of metabolic disease. We use these findings to uncover SFXN5, LETMD1, and ATP1A2 as modulators of BAT thermogenesis or adiposity, and provide OPABAT as a resource for understanding the conserved mechanisms of BAT regulation over metabolic physiology.

    View details for DOI 10.1016/j.cell.2022.10.003

    View details for PubMedID 36334589

  • Metabolic reconstitution of germ-free mice by a gnotobiotic microbiota varies over the circadian cycle. PLoS biology Hoces, D., Lan, J., Sun, W., Geiser, T., Staubli, M. L., Cappio Barazzone, E., Arnoldini, M., Challa, T. D., Klug, M., Kellenberger, A., Nowok, S., Faccin, E., Macpherson, A. J., Stecher, B., Sunagawa, S., Zenobi, R., Hardt, W., Wolfrum, C., Slack, E. 2022; 20 (9): e3001743

    Abstract

    The capacity of the intestinal microbiota to degrade otherwise indigestible diet components is known to greatly improve the recovery of energy from food. This has led to the hypothesis that increased digestive efficiency may underlie the contribution of the microbiota to obesity. OligoMM12-colonized gnotobiotic mice have a consistently higher fat mass than germ-free (GF) or fully colonized counterparts. We therefore investigated their food intake, digestion efficiency, energy expenditure, and respiratory quotient using a novel isolator-housed metabolic cage system, which allows long-term measurements without contamination risk. This demonstrated that microbiota-released calories are perfectly balanced by decreased food intake in fully colonized versus gnotobiotic OligoMM12 and GF mice fed a standard chow diet, i.e., microbiota-released calories can in fact be well integrated into appetite control. We also observed no significant difference in energy expenditure after normalization by lean mass between the different microbiota groups, suggesting that cumulative small differences in energy balance, or altered energy storage, must underlie fat accumulation in OligoMM12 mice. Consistent with altered energy storage, major differences were observed in the type of respiratory substrates used in metabolism over the circadian cycle: In GF mice, the respiratory exchange ratio (RER) was consistently lower than that of fully colonized mice at all times of day, indicative of more reliance on fat and less on glucose metabolism. Intriguingly, the RER of OligoMM12-colonized gnotobiotic mice phenocopied fully colonized mice during the dark (active/eating) phase but phenocopied GF mice during the light (fasting/resting) phase. Further, OligoMM12-colonized mice showed a GF-like drop in liver glycogen storage during the light phase and both liver and plasma metabolomes of OligoMM12 mice clustered closely with GF mice. This implies the existence of microbiota functions that are required to maintain normal host metabolism during the resting/fasting phase of circadian cycle and which are absent in the OligoMM12 consortium.

    View details for DOI 10.1371/journal.pbio.3001743

    View details for PubMedID 36126044

  • Remission of obesity and insulin resistance is not sufficient to restore mitochondrial homeostasis in visceral adipose tissue. Redox biology Gonzalez-Franquesa, A., Gama-Perez, P., Kulis, M., Szczepanowska, K., Dahdah, N., Moreno-Gomez, S., Latorre-Pellicer, A., Fernandez-Ruiz, R., Aguilar-Mogas, A., Hoffman, A., Monelli, E., Samino, S., Miro-Blanch, J., Oemer, G., Duran, X., Sanchez-Rebordelo, E., Schneeberger, M., Obach, M., Montane, J., Castellano, G., Chapaprieta, V., Sun, W., Navarro, L., Prieto, I., Castano, C., Novials, A., Gomis, R., Monsalve, M., Claret, M., Graupera, M., Soria, G., Wolfrum, C., Vendrell, J., Fernandez-Veledo, S., Enriquez, J. A., Carracedo, A., Perales, J. C., Nogueiras, R., Herrero, L., Trifunovic, A., Keller, M. A., Yanes, O., Sales-Pardo, M., Guimera, R., Bluher, M., Martin-Subero, J. I., Garcia-Roves, P. M. 2022; 54: 102353

    Abstract

    Metabolic plasticity is the ability of a biological system to adapt its metabolic phenotype to different environmental stressors. We used a whole-body and tissue-specific phenotypic, functional, proteomic, metabolomic and transcriptomic approach to systematically assess metabolic plasticity in diet-induced obese mice after a combined nutritional and exercise intervention. Although most obesity and overnutrition-related pathological features were successfully reverted, we observed a high degree of metabolic dysfunction in visceral white adipose tissue, characterized by abnormal mitochondrial morphology and functionality. Despite two sequential therapeutic interventions and an apparent global healthy phenotype, obesity triggered a cascade of events in visceral adipose tissue progressing from mitochondrial metabolic and proteostatic alterations to widespread cellular stress, which compromises its biosynthetic and recycling capacity. In humans, weight loss after bariatric surgery showed a transcriptional signature in visceral adipose tissue similar to our mouse model of obesity reversion. Overall, our data indicate that obesity prompts a lasting metabolic fingerprint that leads to a progressive breakdown of metabolic plasticity in visceral adipose tissue.

    View details for DOI 10.1016/j.redox.2022.102353

    View details for PubMedID 35777200

  • GPR180 is a component of TGF beta signalling that promotes thermogenic adipocyte function and mediates the metabolic effects of the adipocyte-secreted factor CTHRC1 NATURE COMMUNICATIONS Balazova, L., Balaz, M., Horvath, C., Horvath, A., Moser, C., Kovanicova, Z., Ghosh, A., Ghoshdastider, U., Efthymiou, V., Kiehlmann, E., Sun, W., Dong, H., Ding, L., Amri, E., Nuutila, P., Virtanen, K. A., Niemi, T., Ukropcova, B., Ukropec, J., Pelczar, P., Lamla, T., Hamilton, B., Neubauer, H., Wolfrum, C. 2021; 12 (1): 7144

    Abstract

    Activation of thermogenic brown and beige adipocytes is considered as a strategy to improve metabolic control. Here, we identify GPR180 as a receptor regulating brown and beige adipocyte function and whole-body glucose homeostasis, whose expression in humans is associated with improved metabolic control. We demonstrate that GPR180 is not a GPCR but a component of the TGFβ signalling pathway and regulates the activity of the TGFβ receptor complex through SMAD3 phosphorylation. In addition, using genetic and pharmacological tools, we provide evidence that GPR180 is required to manifest Collagen triple helix repeat containing 1 (CTHRC1) action to regulate brown and beige adipocyte activity and glucose homeostasis. In this work, we show that CTHRC1/GPR180 signalling integrates into the TGFβ signalling as an alternative axis to fine-tune and achieve low-grade activation of the pathway to prevent pathophysiological response while contributing to control of glucose and energy metabolism.

    View details for DOI 10.1038/s41467-021-27442-x

    View details for Web of Science ID 000728313100027

    View details for PubMedID 34880217

  • Lipolysis drives expression of the constitutively active receptor GPR3 to induce adipose thermogenesis CELL Johansen, O., Ma, T., Hansen, J., Markussen, L., Schreiber, R., Reverte-Salisa, L., Dong, H., Christensen, D., Sun, W., Gnad, T., Karavaeva, I., Nielsen, T., Kooijman, S., Cero, C., Dmytriyeva, O., Shen, Y., Razzoli, M., O'Brien, S. L., Kuipers, E. N., Nielsen, C., Orchard, W., Willemsen, N., Jespersen, N., Lundh, M., Sustarsic, E., Hallgren, C., Frost, M., McGonigle, S., Isidor, M., Broholm, C., Pedersen, O., Hansen, J., Grarup, N., Hansen, T., Kjaer, A., Granneman, J. G., Babu, M., Calebiro, D., Nielsen, S., Ryden, M., Soccio, R., Rensen, P. N., Treebak, J., Schwartz, T., Emanuelli, B., Bartolomucci, A., Pfeifer, A., Zechner, R., Scheele, C., Mandrup, S., Gerhart-Hines, Z. 2021; 184 (13): 3502-+

    Abstract

    Thermogenic adipocytes possess a therapeutically appealing, energy-expending capacity, which is canonically cold-induced by ligand-dependent activation of β-adrenergic G protein-coupled receptors (GPCRs). Here, we uncover an alternate paradigm of GPCR-mediated adipose thermogenesis through the constitutively active receptor, GPR3. We show that the N terminus of GPR3 confers intrinsic signaling activity, resulting in continuous Gs-coupling and cAMP production without an exogenous ligand. Thus, transcriptional induction of Gpr3 represents the regulatory parallel to ligand-binding of conventional GPCRs. Consequently, increasing Gpr3 expression in thermogenic adipocytes is alone sufficient to drive energy expenditure and counteract metabolic disease in mice. Gpr3 transcription is cold-stimulated by a lipolytic signal, and dietary fat potentiates GPR3-dependent thermogenesis to amplify the response to caloric excess. Moreover, we find GPR3 to be an essential, adrenergic-independent regulator of human brown adipocytes. Taken together, our findings reveal a noncanonical mechanism of GPCR control and thermogenic activation through the lipolysis-induced expression of constitutively active GPR3.

    View details for DOI 10.1016/j.cell.2021.04.037

    View details for Web of Science ID 000665547300015

    View details for PubMedID 34048700

    View details for PubMedCentralID PMC8238500

  • Quantification of adipocyte numbers following adipose tissue remodeling CELL REPORTS Moser, C., Straub, L. G., Rachamin, Y., Dapito, D. H., Kulenkampff, E., Ding, L., Sun, W., Modica, S., Balaz, M., Wolfrum, C. 2021; 35 (4): 109023

    Abstract

    To analyze the capacity of white and brown adipose tissue remodeling, we developed two mouse lines to label, quantitatively trace, and ablate white, brown, and brite/beige adipocytes at different ambient temperatures. We show here that the brown adipocytes are recruited first and reach a peak after 1 week of cold stimulation followed by a decline during prolonged cold exposure. On the contrary, brite/beige cell numbers plateau after 3 weeks of cold exposure. At thermoneutrality, brown adipose tissue, in spite of being masked by a white-like morphology, retains its brown-like physiology, as Ucp1+ cells can be recovered immediately upon beta3-adrenergic stimulation. We further demonstrate that the recruitment of Ucp1+ cells in response to cold is driven by existing adipocytes. In contrast, the regeneration of the interscapular brown adipose tissue following ablation of Ucp1+ cells is driven by de novo differentiation.

    View details for DOI 10.1016/j.celrep.2021.109023

    View details for Web of Science ID 000644709600003

    View details for PubMedID 33909996

  • Peroxisomal β-oxidation acts as a sensor for intracellular fatty acids and regulates lipolysis. Nature metabolism Ding, L., Sun, W., Balaz, M., He, A., Klug, M., Wieland, S., Caiazzo, R., Raverdy, V., Pattou, F., Lefebvre, P., Lodhi, I. J., Staels, B., Heim, M., Wolfrum, C. 2021; 3 (12): 1648-1661

    Abstract

    To liberate fatty acids (FAs) from intracellular stores, lipolysis is regulated by the activity of the lipases adipose triglyceride lipase (ATGL), hormone-sensitive lipase and monoacylglycerol lipase. Excessive FA release as a result of uncontrolled lipolysis results in lipotoxicity, which can in turn promote the progression of metabolic disorders. However, whether cells can directly sense FAs to maintain cellular lipid homeostasis is unknown. Here we report a sensing mechanism for cellular FAs based on peroxisomal degradation of FAs and coupled with reactive oxygen species (ROS) production, which in turn regulates FA release by modulating lipolysis. Changes in ROS levels are sensed by PEX2, which modulates ATGL levels through post-translational ubiquitination. We demonstrate the importance of this pathway for non-alcoholic fatty liver disease progression using genetic and pharmacological approaches to alter ROS levels in vivo, which can be utilized to increase hepatic ATGL levels and ameliorate hepatic steatosis. The discovery of this peroxisomal β-oxidation-mediated feedback mechanism, which is conserved in multiple organs, couples the functions of peroxisomes and lipid droplets and might serve as a new way to manipulate lipolysis to treat metabolic disorders.

    View details for DOI 10.1038/s42255-021-00489-2

    View details for PubMedID 34903883

    View details for PubMedCentralID PMC8688145

  • Lysosomal lipoprotein processing in endothelial cells stimulates adipose tissue thermogenic adaptation. Cell metabolism Fischer, A. W., Jaeckstein, M. Y., Gottschling, K., Heine, M., Sass, F., Mangels, N., Schlein, C., Worthmann, A., Bruns, O. T., Yuan, Y., Zhu, H., Chen, O., Ittrich, H., Nilsson, S. K., Stefanicka, P., Ukropec, J., Balaz, M., Dong, H., Sun, W., Reimer, R., Scheja, L., Heeren, J. 2021; 33 (3): 547-564.e7

    Abstract

    In response to cold exposure, thermogenic adipocytes internalize large amounts of fatty acids after lipoprotein lipase-mediated hydrolysis of triglyceride-rich lipoproteins (TRL) in the capillary lumen of brown adipose tissue (BAT) and white adipose tissue (WAT). Here, we show that in cold-exposed mice, vascular endothelial cells in adipose tissues endocytose substantial amounts of entire TRL particles. These lipoproteins subsequently follow the endosomal-lysosomal pathway, where they undergo lysosomal acid lipase (LAL)-mediated processing. Endothelial cell-specific LAL deficiency results in impaired thermogenic capacity as a consequence of reduced recruitment of brown and brite/beige adipocytes. Mechanistically, TRL processing by LAL induces proliferation of endothelial cells and adipocyte precursors via beta-oxidation-dependent production of reactive oxygen species, which in turn stimulates hypoxia-inducible factor-1α-dependent proliferative responses. In conclusion, this study demonstrates a physiological role for TRL particle uptake into BAT and WAT and establishes endothelial lipoprotein processing as an important determinant of adipose tissue remodeling during thermogenic adaptation.

    View details for DOI 10.1016/j.cmet.2020.12.001

    View details for PubMedID 33357458

  • ESRRG and PERM1 Govern Mitochondrial Conversion in Brite/Beige Adipocyte Formation FRONTIERS IN ENDOCRINOLOGY Mueller, S., Perdikari, A., Dapito, D. H., Sun, W., Wollscheid, B., Balaz, M., Wolfrum, C. 2020; 11: 387

    Abstract

    When exposed to cold temperatures, mice increase their thermogenic capacity by an expansion of brown adipose tissue mass and the formation of brite/beige adipocytes in white adipose tissue depots. However, the process of the transcriptional changes underlying the conversion of a phenotypic white to brite/beige adipocytes is only poorly understood. By analyzing transcriptome profiles of inguinal adipocytes during cold exposure and in mouse models with a different propensity to form brite/beige adipocytes, we identified ESRRG and PERM1 as modulators of this process. The production of heat by mitochondrial uncoupled respiration is a key feature of brite/beige compared to white adipocytes and we show here that both candidates are involved in PGC1α transcriptional network to positively regulate mitochondrial capacity. Moreover, we show that an increased expression of ESRRG or PERM1 supports the formation of brown or brite/beige adipocytes in vitro and in vivo. These results reveal that ESRRG and PERM1 are early induced in and important regulators of brite/beige adipocyte formation.

    View details for DOI 10.3389/fendo.2020.00387

    View details for Web of Science ID 000543841400001

    View details for PubMedID 32595605

    View details for PubMedCentralID PMC7304443

  • A Genetic Model to Study the Contribution of Brown and Brite Adipocytes to Metabolism CELL REPORTS Challa, T. D., Dapito, D. H., Kulenkampff, E., Kiehlmann, E., Moser, C., Straub, L., Sun, W., Wolfrum, C. 2020; 30 (10): 3424-+

    Abstract

    UCP1-dependent thermogenesis is studied to define new strategies to ameliorate obesity and type 2 diabetes; however, animal models are mostly limited to germline mutations of UCP1, which can effect adaptive changes in UCP1-independent pathways. We develop an inducible mouse model for the sequential ablation of UCP1+ brown and brite/beige adipocytes in adult mice. We demonstrate that activated brown adipocytes can increase systemic energy expenditure (EE) by 30%, while the contribution of brite/beige UCP1+ cells is <5%. Notably, UCP1+ adipocytes do not contribute to circulating FGF21 levels, either at room temperature or after cold exposure. We demonstrate that the FGF21-mediated effects on EE and glucose homeostasis are partially dependent on the presence of UCP1+ cells, while the effect on weight loss is not. In conclusion, acute UCP1+ cell deletion may be a useful model to study the impact of brown and brite/beige adipocytes on metabolism.

    View details for DOI 10.1016/j.celrep.2020.02.055

    View details for Web of Science ID 000519189700019

    View details for PubMedID 32160547

  • Antioxidants protect against diabetes by improving glucose homeostasis in mouse models of inducible insulin resistance and obesity DIABETOLOGIA Straub, L. G., Efthymiou, V., Grandl, G., Balaz, M., Challa, T., Truscello, L., Horvath, C., Moser, C., Rachamin, Y., Arnold, M., Sun, W., Modica, S., Wolfrum, C. 2019; 62 (11): 2094-2105

    Abstract

    In the context of diabetes, the health benefit of antioxidant treatment has been widely debated. In this study, we investigated the effect of antioxidant treatment during the development of insulin resistance and hyperphagia in obesity and partial lipodystrophy.We studied the role of antioxidants in the regulation of insulin resistance using the tamoxifen-inducible fat-specific insulin receptor knockout (iFIRKO) mouse model, which allowed us to analyse the antioxidant's effect in a time-resolved manner. In addition, leptin-deficient ob/ob mice were used as a hyperphagic, chronically obese and diabetic mouse model to validate the beneficial effect of antioxidants on metabolism.Acute induction of insulin receptor knockout in adipocytes changed the substrate preference to fat before induction of a diabetic phenotype including hyperinsulinaemia and hyperglycaemia. In healthy chow-fed animals as well as in morbidly obese mice, this diabetic phase could be reversed within a few weeks. Furthermore, after the induction of insulin receptor knockout in mature adipocytes, iFIRKO mice were protected from subsequent obesity development through high-fat diet feeding. By genetic tracing we show that the persistent fat mass loss in mice after insulin receptor knockout in adipocytes is not caused by the depletion of adipocytes. Treatment of iFIRKO mice with antioxidants postponed and reduced hyperglycaemia by increasing insulin sensitivity. In ob/ob mice, antioxidants rescued both hyperglycaemia and hyperphagia.We conclude that fat mass reduction through insulin resistance in adipocytes is not reversible. Furthermore, it seems unlikely that adipocytes undergo apoptosis during the process of extreme lipolysis, as a consequence of insulin resistance. Antioxidants have a beneficial health effect not only during the acute phase of diabetes development, but also in a temporary fashion once chronic obesity and diabetes have been established.

    View details for DOI 10.1007/s00125-019-4937-7

    View details for Web of Science ID 000491944900014

    View details for PubMedID 31309261

    View details for PubMedCentralID PMC6805816

  • Human brown adipose tissue is phenocopied by classical brown adipose tissue in physiologically humanized mice NATURE METABOLISM de Jong, J. A., Sun, W., Pires, N. D., Frontini, A., Balaz, M., Jespersen, N. Z., Feizi, A., Petrovic, K., Fischer, A. W., Bokhari, M., Niemi, T., Nuutila, P., Cinti, S., Nielsen, S., Scheele, C., Virtanen, K., Cannon, B., Nedergaard, J., Wolfrum, C., Petrovic, N. 2019; 1 (8): 830-843

    Abstract

    Human and rodent brown adipose tissues (BAT) appear morphologically and molecularly different. Here we compare human BAT with both classical brown and brite/beige adipose tissues of 'physiologically humanized' mice: middle-aged mice living under conditions approaching human thermal and nutritional conditions, that is, prolonged exposure to thermoneutral temperature (approximately 30 °C) and to an energy-rich (high-fat, high-sugar) diet. We find that the morphological, cellular and molecular characteristics (both marker and adipose-selective gene expression) of classical brown fat, but not of brite/beige fat, of these physiologically humanized mice are notably similar to human BAT. We also demonstrate, both in silico and experimentally, that in physiologically humanized mice only classical BAT possesses a high thermogenic potential. These observations suggest that classical rodent BAT is the tissue of choice for translational studies aimed at recruiting human BAT to counteract the development of obesity and its comorbidities.

    View details for DOI 10.1038/s42255-019-0101-4

    View details for Web of Science ID 000500745300010

    View details for PubMedID 32694768

  • Maternal overnutrition programs hedonic and metabolic phenotypes across generations through sperm tsRNAs PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Sarker, G., Sun, W., Rosenkranz, D., Pelczar, P., Opitz, L., Efthymiou, V., Wolfrum, C., Peleg-Raibstein, D. 2019; 116 (21): 10547-10556

    Abstract

    There is a growing body of evidence linking maternal overnutrition to obesity and psychopathology that can be conserved across multiple generations. Recently, we demonstrated in a maternal high-fat diet (HFD; MHFD) mouse model that MHFD induced enhanced hedonic behaviors and obesogenic phenotypes that were conserved across three generations via the paternal lineage, which was independent of sperm methylome changes. Here, we show that sperm tRNA-derived small RNAs (tsRNAs) partly contribute to the transmission of such phenotypes. We observe increased expression of sperm tsRNAs in the F1 male offspring born to HFD-exposed dams. Microinjection of sperm tsRNAs from the F1-HFD male into normal zygotes reproduces obesogenic phenotypes and addictive-like behaviors, such as increased preference of palatable foods and enhanced sensitivity to drugs of abuse in the resultant offspring. The expression of several of the differentially expressed sperm tsRNAs predicted targets such as CHRNA2 and GRIN3A, which have been implicated in addiction pathology, are altered in the mesolimbic reward brain regions of the F1-HFD father and the resultant HFD-tsRNA offspring. Together, our findings demonstrate that sperm tsRNA is a potential vector that contributes to the transmission of MHFD-induced addictive-like behaviors and obesogenic phenotypes across generations, thereby emphasizing its role in diverse pathological outcomes.

    View details for DOI 10.1073/pnas.1820810116

    View details for Web of Science ID 000468403400059

    View details for PubMedID 31061112

    View details for PubMedCentralID PMC6534971

  • Inhibition of Mevalonate Pathway Prevents Adipocyte Browning in Mice and Men by Affecting Protein Prenylation CELL METABOLISM Balaz, M., Becker, A. S., Balazova, L., Straub, L., Mueller, J., Gashi, G., Maushart, C., Sun, W., Dong, H., Moser, C., Horvath, C., Efthymiou, V., Rachamin, Y., Modica, S., Zellweger, C., Bacanovic, S., Stefanicka, P., Varga, L., Ukropcova, B., Profant, M., Opitz, L., Amri, E., Akula, M. K., Bergo, M., Ukropec, J., Falk, C., Zamboni, N., Betz, M., Burger, I. A., Wolfrum, C. 2019; 29 (4): 901-+

    Abstract

    Recent research focusing on brown adipose tissue (BAT) function emphasizes its importance in systemic metabolic homeostasis. We show here that genetic and pharmacological inhibition of the mevalonate pathway leads to reduced human and mouse brown adipocyte function in vitro and impaired adipose tissue browning in vivo. A retrospective analysis of a large patient cohort suggests an inverse correlation between statin use and active BAT in humans, while we show in a prospective clinical trial that fluvastatin reduces thermogenic gene expression in human BAT. We identify geranylgeranyl pyrophosphate as the key mevalonate pathway intermediate driving adipocyte browning in vitro and in vivo, whose effects are mediated by geranylgeranyltransferases (GGTases), enzymes catalyzing geranylgeranylation of small GTP-binding proteins, thereby regulating YAP1/TAZ signaling through F-actin modulation. Conversely, adipocyte-specific ablation of GGTase I leads to impaired adipocyte browning, reduced energy expenditure, and glucose intolerance under obesogenic conditions, highlighting the importance of this pathway in modulating brown adipocyte functionality and systemic metabolism.

    View details for DOI 10.1016/j.cmet.2018.11.017

    View details for Web of Science ID 000463015800013

    View details for PubMedID 30581121

  • BATLAS: Deconvoluting Brown Adipose Tissue CELL REPORTS Perdikari, A., Leparc, G., Balaz, M., Pires, N. D., Lidell, M. E., Sun, W., Fernandez-Albert, F., Mueller, S., Akchiche, N., Dong, H., Balazova, L., Opitz, L., Roder, E., Klein, H., Stefanicka, P., Varga, L., Nuutila, P., Virtanen, K. A., Niemi, T., Taittonen, M., Rudofsky, G., Ukropec, J., Enerback, S., Stupka, E., Neubauer, H., Wolfrum, C. 2018; 25 (3): 784-+

    Abstract

    Recruitment and activation of thermogenic adipocytes have received increasing attention as a strategy to improve systemic metabolic control. The analysis of brown and brite adipocytes is complicated by the complexity of adipose tissue biopsies. Here, we provide an in-depth analysis of pure brown, brite, and white adipocyte transcriptomes. By combining mouse and human transcriptome data, we identify a gene signature that can classify brown and white adipocytes in mice and men. Using a machine-learning-based cell deconvolution approach, we develop an algorithm proficient in calculating the brown adipocyte content in complex human and mouse biopsies. Applying this algorithm, we can show in a human weight loss study that brown adipose tissue (BAT) content is associated with energy expenditure and the propensity to lose weight. This online available tool can be used for in-depth characterization of complex adipose tissue samples and may support the development of therapeutic strategies to increase energy expenditure in humans.

    View details for DOI 10.1016/j.celrep.2018.09.044

    View details for Web of Science ID 000448217500022

    View details for PubMedID 30332656

  • A stromal cell population that inhibits adipogenesis in mammalian fat depots NATURE Schwalie, P. C., Dong, H., Zachara, M., Russeil, J., Alpern, D., Akchiche, N., Caprara, C., Sun, W., Schlaudraff, K., Soldati, G., Wolfrum, C., Deplancke, B. 2018; 559 (7712): 103-+

    Abstract

    Adipocyte development and differentiation have an important role in the aetiology of obesity and its co-morbidities1,2. Although multiple studies have investigated the adipogenic stem and precursor cells that give rise to mature adipocytes3-14, our understanding of their in vivo origin and properties is incomplete2,15,16. This is partially due to the highly heterogeneous and unstructured nature of adipose tissue depots17, which has proven difficult to molecularly dissect using classical approaches such as fluorescence-activated cell sorting and Cre-lox lines based on candidate marker genes16,18. Here, using the resolving power of single-cell transcriptomics19 in a mouse model, we reveal distinct subpopulations of adipose stem and precursor cells in the stromal vascular fraction of subcutaneous adipose tissue. We identify one of these subpopulations as CD142+ adipogenesis-regulatory cells, which can suppress adipocyte formation in vivo and in vitro in a paracrine manner. We show that adipogenesis-regulatory cells are refractory to adipogenesis and that they are functionally conserved in humans. Our findings point to a potentially critical role for adipogenesis-regulatory cells in modulating adipose tissue plasticity, which is linked to metabolic control, differential insulin sensitivity and type 2 diabetes.

    View details for DOI 10.1038/s41586-018-0226-8

    View details for Web of Science ID 000437267400048

    View details for PubMedID 29925944

  • Peroxisome Proliferator Activated Receptor Gamma Controls Mature Brown Adipocyte Inducibility through Glycerol Kinase CELL REPORTS Lasar, D., Rosenwald, M., Kiehlmann, E., Balaz, M., Tall, B., Opitz, L., Lidell, M. E., Zamboni, N., Krznar, P., Sun, W., Varga, L., Stefanicka, P., Ukropec, J., Nuutila, P., Virtanen, K., Amri, E., Enerback, S., Wahli, W., Wolfrum, C. 2018; 22 (3): 760-773

    Abstract

    Peroxisome proliferator-activated receptors (PPARs) have been suggested as the master regulators of adipose tissue formation. However, their role in regulating brown fat functionality has not been resolved. To address this question, we generated mice with inducible brown fat-specific deletions of PPARα, β/δ, and γ, respectively. We found that both PPARα and β/δδ are dispensable for brown fat function. In contrast, we could show that ablation of PPARγ in vitro and in vivo led to a reduced thermogenic capacity accompanied by a loss of inducibility by β-adrenergic signaling, as well as a shift from oxidative fatty acid metabolism to glucose utilization. We identified glycerol kinase (Gyk) as a partial mediator of PPARγ function and could show that Gyk expression correlates with brown fat thermogenic capacity in human brown fat biopsies. Thus, Gyk might constitute the link between PPARγ-mediated regulation of brown fat function and activation by β-adrenergic signaling.

    View details for DOI 10.1016/j.celrep.2017.12.067

    View details for Web of Science ID 000423449400016

    View details for PubMedID 29346772

  • Bmp4 Promotes a Brown to White-like Adipocyte Shift CELL REPORTS Modica, S., Straub, L. G., Balaz, M., Sun, W., Varga, L., Stefanicka, P., Profant, M., Simon, E., Neubauer, H., Ukropcova, B., Ukropec, J., Wolfrum, C. 2016; 16 (8): 2243-2258

    Abstract

    While Bmp4 has a well-established role in the commitment of mesenchymal stem cells into the adipogenic lineage, its role in brown adipocyte formation and activity is not well defined. Here, we show that Bmp4 has a dual function in adipogenesis by inducing adipocyte commitment while inhibiting the acquisition of a brown phenotype during terminal differentiation. Selective brown adipose tissue overexpression of Bmp4 in mice induces a shift from a brown to a white-like adipocyte phenotype. This effect is mediated by Smad signaling and might be in part due to suppression of lipolysis, via regulation of hormone sensitive lipase expression linked to reduced Ppar activity. Given that we observed a strong correlation between BMP4 levels and adipocyte size, as well as insulin sensitivity in humans, we propose that Bmp4 is an important factor in the context of obesity and type 2 diabetes.

    View details for DOI 10.1016/j.celrep.2016.07.048

    View details for Web of Science ID 000382310100020

    View details for PubMedID 27524617

  • Dietary ratios of n-6/n-3 polyunsaturated fatty acids during maternal pregnancy affect hippocampal neurogenesis and apoptosis in mouse offspring NUTRICION HOSPITALARIA Fan, C., Sun, W., Fu, H., Dong, H., Xia, L., Lu, Y., Deckelbaum, R. J., Qi, K. 2015; 32 (3): 1170-1179

    Abstract

    although n-3 polyunsaturated fatty acids (PUFAs) play crucial roles in brain development and function, neither the optimal level of n-3 PUFAs nor the optimal ratio of n-6/n-3 PUFAs in the maternal diet are well defined. In this study, we investigated the effects of dietary n-6/n-3 PUFA ratios during pregnancy on neurogenesis and apoptosis in the brains of mouse offspring. Metods: female C57BL/6J mice were fed one of three diets with high, medium and low ratios of n-6/n-3 PUFAs (15.7:1, 6.3:1, 1.6:1), as well as a high fish oil diet with a n-6/n-3 ratio of 1:5.7; an n-3 PUFA-deficient diet served as control. The feeding regimens began two months before mouse conception and continued for the duration of the pregnancy. The neurogenesis and apoptosis of hippocampal CA3 area in the offspring were detected.compared to the n-3 PUFA-deficient diet, n-3 PUFA-containing diets, particularly those with n-6/n-3 PUFA ratios of 6.3:1 and 1.6:1, significantly increased both phosphorylation of histone H3 at ser 10 (p-H3ser10) and calretinin-positive cells in hippocampus CA3 of the offspring. Furthermore, increased expression of Bcl2 protein, decreased expression of Bax protein, and reduced caspase 3 activity and numbers of TUNEL apoptotic cells were found in the three diets with high, medium and low n-6/n-3 PUFA ratios. However, there were no differences in any of these parameters between the high fish oil diet group and the n-3 PUFA-deficient diet group.these data suggest that a higher intake of n-3 PUFAs with a lower ratio of n-6/n-3 PUFAs of between about 6:1 to 1:1, supplied to mothers during pregnancy, may benefit brain neurogenesis and apoptosis in offspring. However, excessive maternal intake of n-3 PUFAs may exert a negative influence on brain development in the offspring.

    View details for DOI 10.3305/nh.2015.32.3.9259

    View details for Web of Science ID 000362925300028

    View details for PubMedID 26319835