TY - JOUR AB - Lymphangioleiomyomatosis (LAM) is a progressive lung disease with limited treatments, largely due to an incomplete understanding of its pathogenesis. Lymphatic endothelial cells (LECs) invade LAM cell clusters, which include HMB-45-positive epithelioid cells and smooth muscle α-actin-expressing LAM-associated fibroblasts (LAMFs). Recent evidence shows that LAMFs resemble cancer-associated fibroblasts, with LAMF-LEC interactions contributing to disease progression. To explore these mechanisms, we used spatial transcriptomics on LAM lung tissues and identified a gene cluster enriched in kinase signaling pathways linked to myofibroblasts and co-expressed with LEC markers. Kinase arrays revealed elevated PDGFR and FGFR in LAMFs. Using a 3D co-culture spheroid model of primary LAMFs and LECs, we observed increased invasion in LAMF-LEC spheroids compared to non-LAM fibroblasts. Treatment with sorafenib, a multikinase inhibitor, significantly reduced invasion, outperforming Rapamycin. We also confirmed TSC2-deficient renal angiomyolipoma cells (TSC2-null AML) as key VEGF-A secretors, which was suppressed by sorafenib in both TSC2-null AML cells and LAMFs. These findings highlight VEGF-A and bFGF as potential therapeutic targets and suggest multikinase inhibition as a promising strategy for LAM. AU - Koc-Gunel, S.* AU - Liu, E.C.* AU - Gautam, L.K.* AU - Calvert, B.A.* AU - Murthy, S.* AU - Harriott, N.C.* AU - Nawroth, J. AU - Zhou, B.* AU - Krymskaya, V.P.* AU - Ryan, A.L.* C1 - 73281 C2 - 56976 CY - 2015 Manchester Rd, Ann Arbor, Mi 48104 Usa TI - Targeting fibroblast-endothelial interactions in LAM pathogenesis using 3D spheroid models and spatial transcriptomics. JO - JCI insight VL - 10 IS - 6 PB - Amer Soc Clinical Investigation Inc PY - 2025 SN - 2379-3708 ER - TY - JOUR AB - People living with HIV treated during acute infection are the group for whom achieving functional cure appears most viable. Follicular CD8+ T cells could contribute to HIV reservoir clearance by accessing B cell follicles through CXCR5 expression. This study examines peripheral follicular CD8+ T cells using flow cytometry, transcriptome analyses, and functional assays in people treated during acute (n = 37) and chronic (n = 18) infection, as well as in individuals naturally controlling HIV (n = 20) and living without HIV (n = 10). Our results reveal that early, as opposed to late, treatment initiation preserves antiviral effector functions of follicular CD8+ T cells, which are further enhanced by PD1 inhibition. We also identify a correlation between follicular CD8+ T cells and intact proviral HIV DNA levels in acute, but not chronic, infection. Longitudinal transcriptomic analysis of peripheral effector cells after 48 weeks of suppressive therapy indicated traits of recent antigen exposure, suggesting potential recirculation into lymphoid tissue. These findings underscore the pivotal role of follicular CD8+ T cells in anti-HIV responses and support investigating targeted cure strategies, such as anti-PD1 therapy, especially in individuals initiating treatment during acute infection. AU - Rüeger, S.* AU - Gruener, E.* AU - Wang, D.* AU - Abdool, F.S.* AU - Ober, V.* AU - Vallée, T.* AU - Stirner, R.* AU - Conca, R.* AU - Andrä, I.* AU - Rogers, L.* AU - Zahn, R.* AU - Gersbacher, E.* AU - Eger, J.* AU - Pauli, R.* AU - Postel, N.* AU - Spinner, C.D.* AU - Vehreschild, J.J.* AU - Stecher, M.* AU - Nitschko, H.* AU - Eberle, J.* AU - Bogner, J.R.* AU - Seybold, U.* AU - Draenert, R.* AU - Leslie, A.* AU - Kløverpris, H.N.* AU - Geldmacher, C.* AU - Muenchhoff, M.* AU - Held, K. AU - Roider, J.* C1 - 74005 C2 - 57298 CY - 2015 Manchester Rd, Ann Arbor, Mi 48104 Usa TI - Early treatment and PD1 inhibition enhance HIV-specific functionality of follicular CD8+ T cells. JO - JCI insight VL - 10 IS - 7 PB - Amer Soc Clinical Investigation Inc PY - 2025 SN - 2379-3708 ER - TY - JOUR AB - Resistance to chemotherapy of pancreatic ductal adenocarcinoma (PDAC) is largely driven by intratumoral heterogeneity (ITH) due to tumor cell plasticity and clonal diversity. To develop alternative strategies to overcome this defined mechanism of resistance, tools to monitor and quantify ITH in a rapid and scalable fashion are needed urgently. Here, we employed label-free digital holographic microscopy (DHM) to characterize ITH in PDAC. We established a robust experimental and machine learning analysis pipeline to perform single-cell phenotyping based on DHM-derived phase images of PDAC cells in suspension. Importantly, we were able to detect dynamic changes in tumor cell differentiation and heterogeneity of distinct PDAC subtypes upon induction of epithelial-mesenchymal transition and under treatment-imposed pressure in murine and patient-derived model systems. This platform allowed us to assess phenotypic ITH in PDAC on a single-cell level in real time. Implementing this technology into the clinical workflow has the potential to fundamentally increase our understanding of tumor heterogeneity during evolution and treatment response. AU - Wittenzellner, K.* AU - Lengl, M.* AU - Röhrl, S.* AU - Maurer, C.* AU - Klenk, C.* AU - Papargyriou, A. AU - Schmidleitner, L.* AU - Kabella, N.* AU - Shastri, A.R.* AU - Fresacher, D.E.* AU - Harb, F.* AU - Hafez, N.* AU - Bärthel, S.* AU - Lucarelli, D.* AU - Escorial-Iriarte, C.* AU - Orben, F.* AU - Öllinger, R.* AU - Emken, E.* AU - Fricke, L.* AU - Madej, J.* AU - Wustrow, P.* AU - Ekin Demir, I.* AU - Friess, H.* AU - Lahmer, T.* AU - Schmid, R.M.* AU - Rad, R.* AU - Schneider, G.* AU - Kuster, B.* AU - Saur, D.* AU - Hayden, O.* AU - Diepold, K.* AU - Reichert, M. C1 - 75190 C2 - 57828 CY - 2015 Manchester Rd, Ann Arbor, Mi 48104 Usa TI - Label-free single-cell phenotyping to determine tumor cell heterogeneity in pancreatic cancer in real time. JO - JCI insight VL - 10 IS - 13 PB - Amer Soc Clinical Investigation Inc PY - 2025 SN - 2379-3708 ER - TY - JOUR AB - Idiopathic pulmonary fibrosis (IPF) is a lethal chronic lung disease characterized by aberrant intercellular communication, extracellular matrix deposition, and destruction of functional lung tissue. While extracellular vesicles (EVs) accumulate in the IPF lung, their cargo and biological effects remain unclear. We interrogated the proteome of EV and non-EV fractions during pulmonary fibrosis and characterized their contribution to fibrosis. EVs accumulated 14 days after bleomycin challenge, correlating with decreased lung function and initiated fibrogenesis in healthy precision-cut lung slices. Label-free proteomics of bronchoalveolar lavage fluid EVs (BALF-EVs) collected from mice challenged with bleomycin or control identified 107 proteins enriched in fibrotic vesicles. Multiomic analysis revealed fibroblasts as a major cellular source of BALF-EV cargo, which was enriched in secreted frizzled related protein 1 (SFRP1). Sfrp1 deficiency inhibited the activity of fibroblast-derived EVs to potentiate lung fibrosis in vivo. SFRP1 led to increased transitional cell markers, such as keratin 8, and WNT/β-catenin signaling in primary alveolar type 2 cells. SFRP1 was expressed within the IPF lung and localized at the surface of EVs from patient-derived fibroblasts and BALF. Our work reveals altered EV protein cargo in fibrotic EVs promoting fibrogenesis and identifies fibroblast-derived vesicular SFRP1 as a fibrotic mediator and potential therapeutic target for IPF. AU - Burgy, O.* AU - Mayr, C.H.* AU - Schenesse, D.* AU - Fousekis Papakonstantinou, E.* AU - Ballester, B. AU - Sengupta, A. AU - She, Y.* AU - Hu, Q.* AU - Melo-Narváez, M.C AU - Jain, E. AU - Pestoni, J. AU - Mozurak, M.* AU - Estrada-Bernal, A.* AU - Onwuka, U.* AU - Coughlan, C.* AU - Parimon, T.* AU - Chen, P.* AU - Heimerl, T.* AU - Bange, G.* AU - Schmeck, B.T.* AU - Lindner, M. AU - Hilgendorff, A. AU - Ruppert, C.* AU - Guenther, A.* AU - Mann, M.* AU - Yildirim, A.Ö. AU - Eickelberg, O.* AU - Jung, A.L.* AU - Schiller, H. AU - Lehmann, M. AU - Burgstaller, G. AU - Königshoff, M.* C1 - 71807 C2 - 56433 CY - 2015 Manchester Rd, Ann Arbor, Mi 48104 Usa TI - Fibroblast-derived extracellular vesicles contain SFRP1 and mediate pulmonary fibrosis. JO - JCI insight VL - 9 IS - 18 PB - Amer Soc Clinical Investigation Inc PY - 2024 SN - 2379-3708 ER - TY - JOUR AB - Transcriptomic analyses have advanced the understanding of complex disease pathophysiology including chronic obstructive pulmonary disease (COPD). However, identifying relevant biologic causative factors has been limited by the integration of high dimensionality data. COPD is characterized by lung destruction and inflammation with smoke exposure being a major risk factor. To define novel biological mechanisms in COPD, we utilized unsupervised and supervised interpretable machine learning analyses of single cell-RNA sequencing data from the gold standard mouse smoke exposure model to identify significant latent factors (context-specific co-expression modules) impacting pathophysiology. The machine learning transcriptomic signatures coupled to protein networks uncovered a reduction in network complexity and novel biological alterations in actin-associated gelsolin (GSN), which was transcriptionally linked to disease state. GSN was altered in airway epithelial cells in the mouse model and in human COPD. GSN was increased in plasma from COPD patients, and smoke exposure resulted in enhanced GSN release from airway cells from COPD patients. This method provides insights into rewiring of transcriptional networks that are associated with COPD pathogenesis and provide a novel analytical platform for other diseases. AU - Sui, J.* AU - Xiao, H.* AU - Mbaekwe, U.* AU - Ting, N.C.* AU - Murday, K.* AU - Hu, Q.* AU - Gregory, A.D.* AU - Kapellos, T. AU - Yildirim, A.Ö. AU - Königshoff, M.* AU - Zhang, Y.* AU - Sciurba, F.C.* AU - Das, J.* AU - Kliment, C.R.* C1 - 71881 C2 - 56470 TI - Interpretable machine learning uncovers epithelial transcriptional rewiring and a role for Gelsolin in COPD. JO - JCI insight VL - 9 IS - 21 PY - 2024 SN - 2379-3708 ER - TY - JOUR AB - Ovarian cancer (OvCa) preferentially metastasizes in association with mesothelial cell-lined surfaces. We sought to determine if mesothelial cells are required for OvCa metastasis and detect alterations in mesothelial cell gene expression and cytokine secretion upon interaction with OvCa cells. Using omental samples from patients with high-grade serous OvCa and mouse models with Wt1-driven GFP-expressing mesothelial cells, we validated the intratumoral localization of mesothelial cells during human and mouse OvCa omental metastasis. Removing mesothelial cells ex vivo from human and mouse omenta or in vivo using diphtheria toxin mediated ablation in Msln-Cre mice, significantly inhibited OvCa cell adhesion and colonization. Human ascites induced angiopoietin-like 4 (ANGPTL4) and stanniocalcin 1 (STC1) expression and secretion by mesothelial cells. Inhibition of STC1 or ANGPTL4 via RNAi, obstructed OvCa cell-induced mesothelial cell to mesenchymal transition while inhibition of ANGPTL4 alone obstructed OvCa cell-induced mesothelial cell migration and glycolysis. Inhibition of mesothelial cell ANGPTL4 secretion via RNAi prevented mesothelial cell induced monocyte migration, endothelial cell vessel formation and OvCa cell adhesion, migration, and proliferation. In contrast, inhibition of mesothelial cell STC1 secretion via RNAi prevented mesothelial cell induced endothelial cell vessel formation and OvCa cell adhesion, migration, proliferation, and invasion. Additionally, blocking ANPTL4 function with antibodies reduced the ex vivo colonization of three different OvCa cell lines on human omental tissue explants and in vivo colonization of ID8p53-/-Brca2-/- cells on mouse omenta. These findings indicate that mesothelial cells are important to the initial stages of OvCa metastasis, and that the crosstalk between mesothelial cells and the tumor microenvironment, promotes OvCa metastasis through the secretion of ANGPTL4. AU - Bajwa, P.* AU - Kordylewicz, K.* AU - Bilecz, A.* AU - Lastra, R.R.* AU - Wroblewski, K.* AU - Rinkevich, Y. AU - Lengyel, E.* AU - Kenny, H.A.* C1 - 67468 C2 - 54123 CY - 2015 Manchester Rd, Ann Arbor, Mi 48104 Usa TI - Cancer-associated mesothelial cell-derived ANGPTL4 and STC1 promote the early steps of ovarian cancer metastasis. JO - JCI insight VL - 8 IS - 6 PB - Amer Soc Clinical Investigation Inc PY - 2023 SN - 2379-3708 ER - TY - JOUR AB - Most overweight individuals do not develop diabetes due to compensatory islet responses to restore glucose homeostasis. Therefore, regulatory pathways that promote β-cell compensation are potential targets for treatment of diabetes. The melastatin transient receptor potential 7 protein (TRPM7), harboring a cation channel and a serine/threonine kinase, has been implicated in controlling cell growth and proliferation. Here, we report that selective deletion of Trpm7 in β-cells disrupts insulin secretion and leads to progressive glucose intolerance. We indicate that the diminished insulinotropic response in β-cell-specific Trpm7 knockout mice is caused by decreased insulin production due to an impaired enzymatic activity of this protein. Accordingly, high-fat fed mice with a genetic loss of TRPM7 kinase activity (Trpm7R/R) display a marked glucose intolerance accompanied by hyperglycemia. These detrimental glucoregulatory effects are engendered by reduced compensatory β-cell responses due to mitigated AKT/ERK signaling. Collectively, our data identify TRPM7 kinase as a novel regulator of insulin synthesis, β-cell dynamics, and glucose homeostasis under obesogenic diet. AU - Khajavi, N.* AU - Beck, A.* AU - Ricku, K.* AU - Beyerle, P.* AU - Jacob, K.* AU - Syamsul, S.F.* AU - Belkacemi, A.* AU - Reinach, P.S.* AU - Schreier, P.C.* AU - Salah, H.* AU - Popp, T.* AU - Novikoff, A. AU - Breit, A.* AU - Chubanov, V.* AU - Müller, T.D. AU - Zierler, S.* AU - Gudermann, T.* C1 - 67128 C2 - 53444 CY - 2015 Manchester Rd, Ann Arbor, Mi 48104 Usa TI - TRPM7 kinase is required for insulin production and compensatory islet responses during obesity. JO - JCI insight VL - 8 IS - 3 PB - Amer Soc Clinical Investigation Inc PY - 2023 SN - 2379-3708 ER - TY - JOUR AB - Newborns are at high risk of developing neonatal sepsis, particularly if born prematurely. This has been linked to divergent requirements the immune system has to fulfill during intrauterine compared to extrauterine life. By transcriptomic analysis of fetal and adult neutrophils we set out to shed new light on the molecular mechanisms of neutrophil maturation and functional adaption during fetal ontogeny. We identified an accumulation of differentially regulated genes within the non-canonical NF-κB signaling pathway accompanied by constitutive nuclear localization of RelB and increased surface expression of TNFRII in fetal neutrophils as well as elevated levels of LT-α in fetal serum. Furthermore, we found strong upregulation of the negative inflammatory regulator A20 (Tnfaip3) in fetal neutrophils, which was accompanied by a pronounced downregulation of the canonical NF-κB pathway. Functionally, overexpressing A20 in Hoxb8 cells led to reduced adhesion of these neutrophil-like cells under flow. Conversely, mice with a neutrophil specific A20 deletion displayed increased inflammation in vivo. Taken together, we have uncovered constitutive activation of the non-canonical NF-κB pathway with concomitant upregulation of A20 in fetal neutrophils. This offers perfect adaption of neutrophil function during intrauterine fetal life, but also restricts appropriate immune responses particularly in prematurely born infants. AU - Rohwedder, I.* AU - Wackerbarth, L.M.* AU - Heinig, K.* AU - Ballweg, A.* AU - Altstätter, J.* AU - Ripphahn, M.* AU - Nussbaum, C.* AU - Salvermoser, M.* AU - Bierschenk, S.* AU - Straub, T.* AU - Gunzer, M.* AU - Schmidt-Supprian, M.* AU - Kolben, T.* AU - Schulz, C.* AU - Ma, A.* AU - Walzog, B.* AU - Heinig, M. AU - Sperandio, M.* C1 - 67208 C2 - 54219 TI - A20 and the non-canonical NF-κB pathway are key regulators of neutrophil recruitment during fetal ontogeny. JO - JCI insight VL - 8 IS - 4 PY - 2023 SN - 2379-3708 ER - TY - JOUR AB - Spatially resolved metabolomics enables the investigation of tumoral metabolites in situ. Inter- and intratumor heterogeneity are key factors associated with patient outcomes. Adrenocortical carcinoma (ACC) is an exceedingly rare tumor associated with poor survival. Its clinical prognosis is highly variable, but the contributions of tumor metabolic heterogeneity have not been investigated thus far to our knowledge. An in-depth understanding of tumor heterogeneity requires molecular feature-based identification of tumor subpopulations associated with tumor aggressiveness. Here, using spatial metabolomics by high-mass resolution MALDI Fourier transform ion cyclotron resonance mass spectrometry imaging, we assessed metabolic heterogeneity by de novo discovery of metabolic subpopulations and Simpson's diversity index. After identification of tumor subpopulations in 72 patients with ACC, we additionally performed a comparison with 25 tissue sections of normal adrenal cortex to identify their common and unique metabolic subpopulations. We observed variability of ACC tumor heterogeneity and correlation of high metabolic heterogeneity with worse clinical outcome. Moreover, we identified tumor subpopulations that served as independent prognostic factors and, furthermore, discovered 4 associated anticancer drug action pathways. Our research may facilitate comprehensive understanding of the biological implications of tumor subpopulations in ACC and showed that metabolic heterogeneity might impact chemotherapy. AU - Wang, Q. AU - Sun, N. AU - Meixner, R. AU - Le Gleut, R. AU - Kunzke, T. AU - Feuchtinger, A. AU - Wang, J. AU - Shen, J. AU - Kircher, S.* AU - Dischinger, U.* AU - Weigand, I.* AU - Beuschlein, F.* AU - Fassnacht, M.* AU - Kroiss, M.* AU - Walch, A.K. C1 - 67973 C2 - 54451 CY - 2015 Manchester Rd, Ann Arbor, Mi 48104 Usa TI - Metabolic heterogeneity in adrenocortical carcinoma impacts patient outcomes. JO - JCI insight VL - 8 IS - 16 PB - Amer Soc Clinical Investigation Inc PY - 2023 SN - 2379-3708 ER - TY - JOUR AB - Patients with the renal phosphate-wasting disease X-linked hypophosphatemia (XLH) and Hyp mice, the murine homolog of XLH, are characterized by loss-of-function mutations in phosphate-regulating endopeptidase homolog X-linked (PHEX), leading to excessive secretion of the bone-derived phosphotropic hormone FGF23. The mineralization defect in patients with XLH and Hyp mice is caused by a combination of hypophosphatemia and local accumulation of mineralization-inhibiting molecules in bone. However, the mechanism by which PHEX deficiency regulates bone cell metabolism remains elusive. Here, we used spatial metabolomics by employing matrix-assisted laser desorption/ionization (MALDI) Fourier-transform ion cyclotron resonance mass spectrometry imaging (MSI) of undecalcified bone cryosections to characterize in situ metabolic changes in bones of Hyp mice in a holistic, unbiased manner. We found complex changes in Hyp bone metabolism, including perturbations in pentose phosphate, purine, pyrimidine, and phospholipid metabolism. Importantly, our study identified an upregulation of several biochemical pathways involved in intra- and extracellular production of the mineralization inhibitor pyrophosphate in the bone matrix of Hyp mice. Our data emphasize the utility of MSI-based spatial metabolomics in bone research and provide holistic in situ insights as to how Phex deficiency-induced changes in biochemical pathways in bone cells are linked to impaired bone mineralization. AU - Buck, A. AU - Prade, V.M. AU - Kunzke, T. AU - Erben, R.G.* AU - Walch, A.K. C1 - 66533 C2 - 53206 TI - Spatial metabolomics reveals upregulation of several pyrophosphate-producing pathways in cortical bone of Hyp mice. JO - JCI insight VL - 7 IS - 20 PY - 2022 SN - 2379-3708 ER - TY - JOUR AB - The molecular mechanisms that drive the acquisition of distinct neural crest cell (NCC) fates is still poorly understood. Here, we identify Prdm6 as an epigenetic modifier that temporally and spatially regulates the expression of NCC specifiers and determines the fate of a subset of migrating Cardiac NCCs (CNCCs). Using transcriptomic analysis, genetic and fate mapping approaches in transgenic mice, we show that disruption of Prdm6 is associated with impaired CNCC differentiation, delamination, and migration, and leads to patent ductus arteriosus (DA)and ventricular noncompaction. Bulk and single-cell RNA-seq analyses of DA and CNCC identify Prdm6 as a regulator of a network of CNCC specification genes including Wnt1, Tfap2b, and Sox9. Loss of Prdm6 in CNCCs diminishes its expression in pre-EMT cluster, resulting in the retention of NCC in the dorsal neural tube. This defect is associated with diminished H4K20 mono-methylation and G1-S progression and augmented Wnt1 transcript levels in pre-EMT and neural tube clusters, which we show is the major driver of the impaired CNCC migration. Altogether, these findings reveal Prdm6 as a key regulator of CNCC differentiation and migration and identify Prdm6 and its regulated network as potential targets for the treatment of congenital heart diseases. AU - Hong, L.* AU - Li, N.* AU - Gasque, V.* AU - Mehta, S.* AU - Ye, L.* AU - Wu, Y.* AU - Li, J.* AU - Gewies, A. AU - Ruland, J.* AU - Hirschi, K.K.* AU - Eichmann, A.* AU - Hendry, C.* AU - van Dijk, D.* AU - Mani, A.* C1 - 64252 C2 - 52159 CY - 2015 Manchester Rd, Ann Arbor, Mi 48104 Usa TI - Prdm6 controls heart development by regulating neural crest cell differentiation and migration. JO - JCI insight VL - 7 IS - 4 PB - Amer Soc Clinical Investigation Inc PY - 2022 SN - 2379-3708 ER - TY - JOUR AB - BACKGROUNDInsulin resistance of the brain can unfavorably affect long-term weight maintenance and body fat distribution. Little is known if and how brain insulin sensitivity can be restored in humans. We aimed to evaluate the effects of an exercise intervention on insulin sensitivity of the brain and how this relates to exercise-induced changes in whole-body metabolism and behavior.METHODSIn this clinical trial, sedentary participants who were overweight and obese underwent an 8-week supervised aerobic training intervention. Brain insulin sensitivity was assessed in 21 participants (14 women, 7 men; age range 21-59 years; BMI range 27.5-45.5 kg/m2) using functional MRI, combined with intranasal administration of insulin, before and after the intervention.RESULTSThe exercise program resulted in enhanced brain insulin action to the level of a person of healthy weight, demonstrated by increased insulin-induced striatal activity and strengthened hippocampal functional connectivity. Improved brain insulin action correlated with increased mitochondrial respiration in skeletal muscle, reductions in visceral fat and hunger, as well as improved cognition. Mediation analyses suggest that improved brain insulin responsiveness helps mediate the peripheral exercise effects leading to healthier body fat distribution and reduced perception of hunger.CONCLUSIONOur study demonstrates that an 8-week exercise intervention in sedentary individuals can restore insulin action in the brain. Hence, the ameliorating benefits of exercise toward brain insulin resistance may provide an objective therapeutic target in humans in the challenge to reduce diabetes risk factors.TRIAL REGISTRATIONClinicalTrials.gov (NCT03151590).FUNDINGBMBF/DZD 01GI0925. AU - Kullmann, S. AU - Goj, T. AU - Veit, R. AU - Fritsche, L. AU - Wagner, L. AU - Schneeweiss, P.* AU - Hoene, M.* AU - Hoffmann, C.* AU - Machann, J. AU - Niess, A.* AU - Preissl, H. AU - Birkenfeld, A.L. AU - Peter, A. AU - Häring, H.-U. AU - Fritsche, A. AU - Moller, A. AU - Weigert, C. AU - Heni, M. C1 - 66252 C2 - 52753 TI - Exercise restores brain insulin sensitivity in sedentary adults who are overweight and obese. JO - JCI insight VL - 7 IS - 18 PY - 2022 SN - 2379-3708 ER - TY - JOUR AB - UMOD is a major risk gene for monogenic and complex forms of kidney disease. The encoded kidney-specific protein uromodulin is highly abundant in urine and related to chronic kidney disease, hypertension, and pathogen defense. To gain insights into potential systemic roles, we performed genome-wide screens of circulating uromodulin using complementary antibody-based (N=13,985) and aptamer-based (N=18,070) assays. We detected 3 and 10 distinct significant (p<5e-8) loci, respectively. Integration of antibody-based results at the UMOD locus with functional genomics data (RNA-seq, ATAC-seq, Hi-C) of primary human kidney tissue highlights an upstream variant with differential accessibility and transcription in uromodulin-synthesizing kidney cells as underlying the observed cis effect. Shared association patterns with complex traits, including chronic kidney disease and blood pressure, place the PRKAG2 locus in the same pathway as UMOD. Experimental validation of the third antibody-based locus, B4GALNT2, shows that the p.Cys466Arg variant of the encoded N-acetylgalactosaminyltransferase has a loss-of-function effect leading to higher serum uromodulin levels. Aptamer-based results point to enzymes writing glycan marks present on uromodulin and to their receptors in the circulation, suggesting that this assay permits investigating uromodulin's complex glycosylation rather than its quantitative levels. Overall, our study provides new insights into circulating uromodulin and its emerging functions. AU - Li, Y.* AU - Cheng, Y.* AU - Consolato, F.* AU - Schiano, G.* AU - Chong, M.R.* AU - Pietzner, M.* AU - Nguyen, N.Q.H.* AU - Scherer, N.* AU - Biggs, M.L.* AU - Kleber, M.E.* AU - Haug, S.* AU - Göçmen, B.* AU - Pigeyre, M.* AU - Sekula, P.* AU - Steinbrenner, I.* AU - Schlosser, P.* AU - Joseph, C.B.* AU - Brody, J.A.* AU - Grams, M.E.* AU - Hayward, C.* AU - Schultheiss, U.T.* AU - Krämer, B.K.* AU - Kronenberg, F.* AU - Peters, A. AU - Seissler, J.* AU - Steubl, D.* AU - Then, C.* AU - Wuttke, M.* AU - März, W.* AU - Eckardt, K.U.* AU - Gieger, C. AU - Boerwinkle, E.* AU - Psaty, B.M.* AU - Coresh, J.* AU - Oefner, P.J.* AU - Paré, G.* AU - Langenberg, C.* AU - Scherberich, J.E.* AU - Yu, B.* AU - Akilesh, S.* AU - Devuyst, O.* AU - Rampoldi, L.* AU - Köttgen, A.* C1 - 64888 C2 - 52236 TI - Genome-wide studies reveal factors associated with circulating uromodulin and its relations with complex diseases. JO - JCI insight VL - 7 IS - 10 PY - 2022 SN - 2379-3708 ER - TY - JOUR AB - In many solid cancers, tumor-associated macrophages (TAM) represent the predominant myeloid cell population. Antigen (Ag) cross-presentation leading to tumor Ag-directed cytotoxic CD8+ T cell responses is crucial for anti-tumor immunity. However, the role of recruited monocyte-derived macrophages, including TAM, as potential cross-presenting cells is not well understood. Here, we show that primary human as well as mouse CD206+ macrophages are effective in functional cross-presentation of soluble self and non-self Ag, including tumor-associated Ag (TAA) as well as viral Ag. To confirm the presence of cross-presenting TAM in vivo, we performed phenotypic and functional analysis of TAM from B16-F10 and CT26 syngeneic tumor models and have identified CD11b+F4/80hiCD206+ TAM to effectively cross-present TAA. We show that CD11b+CD206+ TAM represent the dominant tumor-infiltrating myeloid cell population, expressing a unique cell surface repertoire, promoting Ag cross-presentation and Ag-specific CD8+ T cell activation comparable to cross-presenting CLEC9A+ dendritic cells (cDC1). The presence of cross-presenting CD206+ TAM is associated with reduced tumor burden in mouse syngeneic tumor models and with improved overall survival in cutaneous melanoma patients. Therefore, the demonstration of effective Ag cross-presentation capabilities of CD206+ TAM, including their clinical relevance, expands our understanding of TAM phenotypic diversity and functional versatility. AU - Modak, M.* AU - Mattes, A.K.* AU - Reiss, D.* AU - Skronska-Wasek, W.* AU - Langlois, R.* AU - Sabarth, N.* AU - Konopitzky, R.* AU - Ramírez, F.* AU - Lehr, K.* AU - Mayr, T.* AU - Kind, D.* AU - Viollet, C.* AU - Swee, L.K.* AU - Petschenka, J.* AU - El Kasmi, K.C.* AU - Nößner, E. AU - Kitt, K.* AU - Pflanz, S.* C1 - 65029 C2 - 52649 TI - CD206+ tumor-associated macrophages cross-present tumor antigen and drive anti-tumor immunity. JO - JCI insight VL - 7 IS - 11 PY - 2022 SN - 2379-3708 ER - TY - JOUR AB - Obesity is one of the main drivers of type 2 diabetes, but it is not uniformly associated with the disease. The location of fat accumulation is critical for metabolic health. Specific patterns of body fat distribution, such as visceral fat, are closely related to insulin resistance. There might be further, hitherto unknown, features of body fat distribution that could additionally contribute to the disease. We used machine learning with dense convolutional neural networks to detect diabetes-related variables from 2371 T1-weighted whole-body MRI data sets. MRI was performed in participants undergoing metabolic screening with oral glucose tolerance tests. Models were trained for sex, age, BMI, insulin sensitivity, HbA1c, and prediabetes or incident diabetes. The results were compared with those of conventional models. The area under the receiver operating characteristic curve was 87% for the type 2 diabetes discrimination and 68% for prediabetes, both superior to conventional models. Mean absolute regression errors were comparable to those of conventional models. Heatmaps showed that lower visceral abdominal regions were critical in diabetes classification. Subphenotyping revealed a group with high future diabetes and microalbuminuria risk. Our results show that diabetes is detectable from whole-body MRI without additional data. Our technique of heatmap visualization identifies plausible anatomical regions and highlights the leading role of fat accumulation in the lower abdomen in diabetes pathogenesis. AU - Dietz, B.* AU - Machann, J. AU - Agrawal, V.* AU - Heni, M. AU - Schwab, P.* AU - Dienes, J.K.* AU - Reichert, S.* AU - Birkenfeld, A.L. AU - Häring, H.-U. AU - Schick, F. AU - Stefan, N. AU - Fritsche, A. AU - Preissl, H. AU - Schölkopf, B.* AU - Bauer, S.* AU - Wagner, R. C1 - 63523 C2 - 51577 CY - 2015 Manchester Rd, Ann Arbor, Mi 48104 Usa TI - Detection of diabetes from whole-body MRI using deep learning. JO - JCI insight VL - 6 IS - 21 PB - Amer Soc Clinical Investigation Inc PY - 2021 SN - 2379-3708 ER - TY - JOUR AB - Neutrophils provide a critical line of defense in immune responses to various pathogens, but also inflict self-damage upon transition to a hyperactivated, procoagulant state. Recent work has highlighted proinflammatory neutrophil phenotypes contributing to lung injury and acute respiratory distress syndrome (ARDS) in patients suffering from COVID-19. Here, we utilize state-of-the art mass spectrometry-based proteomics, transcriptomic and correlative analyses as well as functional in vitro and in vivo studies to dissect how neutrophils contribute to the progression to severe COVID-19. We identify a reinforcing loop of both systemic and neutrophil intrinsic interleukin-8 (CXCL8/IL-8) dysregulation, which initiates and perpetuates neutrophil-driven immunopathology. This positive feedback loop of systemic and neutrophil autocrine IL-8 production leads to an activated, prothrombotic neutrophil phenotype characterized by degranulation and neutrophil extracellular trap (NET) formation. In severe COVID-19, neutrophils directly initiate the coagulation and complement cascade, highlighting a link to the immunothrombotic state observed in these patients. Targeting the IL-8-CXCR-1/-2 axis interferes with this vicious cycle and attenuates neutrophil activation, degranulation, NETosis, and IL-8 release. Finally, we show that blocking IL-8-like signaling reduces SARS-CoV-2 spike protein-induced, hACE2-dependent pulmonary microthrombosis in mice. In summary, our data provide comprehensive insights into the activation mechanisms of neutrophils in COVID-19 and uncover a self-sustaining neutrophil-IL-8-axis as promising therapeutic target in severe SARS-CoV-2 infection. AU - Kaiser, R.* AU - Leunig, A.* AU - Pekayvaz, K.* AU - Popp, O.* AU - Joppich, M.* AU - Polewka, V.* AU - Escaig, R.* AU - Anjum, A.* AU - Hoffknecht, M.L.* AU - Gold, C.* AU - Brambs, S.* AU - Engel, A.* AU - Stockhausen, S.* AU - Knottenberg, V.* AU - Titova, A.* AU - Haji, M.* AU - Scherer, C.* AU - Muenchhoff, M.* AU - Hellmuth, J.C.* AU - Saar, K.* AU - Schubert, B. AU - Hilgendorff, A. AU - Schulz, C.* AU - Kääb, S.* AU - Zimmer, R.* AU - Hübner, N.* AU - Massberg, S.* AU - Mertins, P.* AU - Nicolai, L.* AU - Stark, K.* C1 - 62896 C2 - 51150 CY - 2015 Manchester Rd, Ann Arbor, Mi 48104 Usa TI - Self-sustaining interleukin-8 loops drive a prothrombotic neutrophil phenotype in severe COVID-19. JO - JCI insight VL - 6 IS - 18 PB - Amer Soc Clinical Investigation Inc PY - 2021 SN - 2379-3708 ER - TY - JOUR AB - Reduced expression of the plasma membrane citrate transporter INDY (acronym I’m Not Dead, Yet) extends life span in lower organisms. Deletion of the mammalian Indy (mIndy) gene in rodents improves metabolism via mechanisms akin to caloric restriction, known to lower blood pressure (BP) by sympathoadrenal inhibition. We hypothesized that mIndy deletion attenuates sympathoadrenal support of BP. Continuous arterial BP and heart rate (HR) were reduced in mINDY-KO mice. Concomitantly, urinary catecholamine content was lower, and the decreases in BP and HR by mIndy deletion were attenuated after autonomic ganglionic blockade. Catecholamine biosynthesis pathways were reduced in mINDY-KO adrenals using unbiased microarray analysis. Citrate, the main mINDY substrate, increased catecholamine content in pheochromocytoma cells, while pharmacological inhibition of citrate uptake blunted the effect. Our data suggest that deletion of mIndy reduces sympathoadrenal support of BP and HR by attenuating catecholamine biosynthesis. Deletion of mIndy recapitulates beneficial cardiovascular and metabolic responses to caloric restriction, making it an attractive therapeutic target. AU - Willmes, D.M. AU - Daniels, M. AU - Kurzbach, A. AU - Lieske, S.* AU - Bechmann, N.* AU - Schumann, T. AU - Henke, C. AU - El-Agroudy, N.N. AU - da Costa Goncalves, A.C.* AU - Peitzsch, M.* AU - Hofmann, A.* AU - Kanczkowski, W.* AU - Kräker, K.* AU - Müller, D.N.* AU - Morawietz, H.* AU - Deussen, A.* AU - Wagner, M.* AU - El-Armouche, A.* AU - Helfand, S.L.* AU - Bornstein, S.R.* AU - de Cabo, R.* AU - Bernier, M.* AU - Eisenhofer, G.* AU - Tank, J.* AU - Jordan, J.* AU - Birkenfeld, A.L. C1 - 61773 C2 - 50089 TI - The longevity gene mIndy (I’m Not Dead, Yet) affects blood pressure through sympathoadrenal mechanisms. JO - JCI insight VL - 6 IS - 2 PY - 2021 SN - 2379-3708 ER - TY - JOUR AB - Ribosomopathies are congenital disorders caused by mutations in the genes encoding ribosomal and other functionally related proteins. They are characterized by anemia, other hematopoietic and developmental abnormalities, and p53 activation. Ribosome assembly requires coordinated expression of many ribosomal protein (RP) genes; however, the regulation of RP gene expression, especially in hematopoietic stem cells (HSCs), remains poorly understood. MYSM1 is a transcriptional regulator essential for HSC function and hematopoiesis. We established that HSC dysfunction in Mysm1 deficiency is driven by p53; however, the mechanisms of p53 activation remained unclear. Here, we describe the transcriptome of Mysm1-deficient mouse HSCs and identify MYSM1 genome-wide DNA binding sites. We establish a direct role for MYSM1 in RP gene expression and show a reduction in protein synthesis in Mysm1(-/-) HSCs. Loss of p53 in mice fully rescues Mysm1(-/-) anemia phenotype but not RP gene expression, indicating that RP gene dysregulation is a direct outcome of Mysm1 deficiency and an upstream mediator of Mysm1-/phenotypes through p53 activation. We characterize a patient with a homozygous nonsense MYSM1 gene variant, and we demonstrate reduced protein synthesis and increased p53 levels in patient hematopoietic cells. Our work provides insights into the specialized mechanisms regulating RP gene expression in HSCs and establishes a common etiology of MYSM1 deficiency and ribosomopathy syndromes. AU - Belle, J.I.* AU - Wang, H.* AU - Fiore, A.* AU - Petrov, J.C.* AU - Lin, Y.H.* AU - Feng, C.H.* AU - Nguyen, T.T.M.* AU - Tung, J.* AU - Campeau, P.M.* AU - Behrends, U.* AU - Brunet, T.* AU - Leszinski, G.S. AU - Gros, P.* AU - Langlais, D.* AU - Nijnik, A.* C1 - 59649 C2 - 48971 CY - 2015 Manchester Rd, Ann Arbor, Mi 48104 Usa TI - MYSM1 maintains ribosomal protein gene expression in hematopoietic stem cells to prevent hematopoietic dysfunction. JO - JCI insight VL - 5 IS - 13 PB - Amer Soc Clinical Investigation Inc PY - 2020 SN - 2379-3708 ER - TY - JOUR AB - In type 1 diabetes (T1D), autoimmune destruction of pancreatic beta cells leads to insulin deficiency and loss of glycemic control. However, knowledge about human pancreas pathophysiology in T1D remains incomplete. To address this limitation, we established a pancreas tissue slice platform of donor organs with and without diabetes, facilitating the first live cell studies of human pancreas in T1D pathogenesis to our knowledge. We show that pancreas tissue slices from organ donors allow thorough assessment of processes critical for disease development, including insulin secretion, beta cell physiology, endocrine cell morphology, and immune infiltration within the same donor organ. Using this approach, we compared detailed pathophysiological profiles for 4 pancreata from donors with T1D with 19 nondiabetic control donors. We demonstrate that cell loss, beta cell dysfunction, alterations of beta cell physiology, and islet infiltration contributed differently to individual cases of T1D, allowing insight into pathophysiology and heterogeneity of T1D pathogenesis. Thus, our study demonstrates that organ donor pancreas tissue slices represent a promising and potentially novel approach in the search for successful prevention and reversal strategies of T1D. AU - Panzer, J.K. AU - Hiller, H.* AU - Cohrs, C.M. AU - Almaça, J.* AU - Enos, S.J. AU - Beery, M.* AU - Cechin, S.* AU - Drotar, D.M. AU - Weitz, J.R.* AU - Santini, J.* AU - Huber, M.K.* AU - Muhammad Fahd Qadir, M.* AU - Pastori, R.L.* AU - Domínguez-Bendala, J.* AU - Phelps, E.A.* AU - Atkinson, M.A.* AU - Pugliese, A.* AU - Caicedo, A.* AU - Kusmartseva, I.* AU - Speier, S. C1 - 58929 C2 - 48474 CY - 2015 Manchester Rd, Ann Arbor, Mi 48104 Usa TI - Pancreas tissue slices from organ donors enable in situ analysis of type 1 diabetes pathogenesis. JO - JCI insight VL - 5 IS - 8 PB - Amer Soc Clinical Investigation Inc PY - 2020 SN - 2379-3708 ER - TY - JOUR AB - Ischemia/reperfusion-induced edema (IRE), one of the most significant causes of mortality after lung transplantation, can be mimicked ex vivo in isolated perfused mouse lungs (IPL). Transient receptor potential vanilloid 4 (TRPV4) is a nonselective cation channel studied in endothelium; however, its role in the lung epithelium remains elusive. Here, we show enhanced IRE in TRPV4-deficient (TRPV4(-/-)) IPL compared with that of WT controls, indicating a protective role of TRPV4 in maintenance of the alveolar epithelial barrier. By immunohistochemistry, mRNA profiling, and electrophysiological characterization, we detected TRPV4 in bronchial epithelium, alveolar epithelial type I (ATI), and alveolar epithelial type II (ATII) cells. Genetic ablation of TRPV4 resulted in reduced expression of the water-conducting aqua porin-5 (AQP-5) channel in ATI cells. Migration of TRPV4(-)(/-) ATI cells was reduced, and cell barrier function was impaired. Analysis of isolated primary TRPV4(-/-) ATII cells revealed a reduced expression of surfactant protein C, and the TRPV4 activator GSK1016790A induced increases in current densities only in WT ATII cells. Moreover, TRPV4(-/-) lungs of adult mice developed significantly larger mean chord lengths and altered lung function compared with WT lungs. Therefore, our data illustrate essential functions of TRPV4 channels in alveolar epithelial cells and in protection from edema formation. AU - Weber, J.* AU - Rajan, S.* AU - Schremmer, C.* AU - Chao, Y.K.* AU - Krasteva-Christ, G.* AU - Kannler, M.* AU - Yildirim, A.Ö. AU - Brosien, M.* AU - Schredelseker, J.* AU - Weissmann, N.* AU - Grimm, C.* AU - Gudermann, T.* AU - Dietrich, A.* C1 - 60373 C2 - 49308 CY - 2015 Manchester Rd, Ann Arbor, Mi 48104 Usa TI - TRPV4 channels are essential for alveolar epithelial barrier function as protection from lung edema. JO - JCI insight VL - 5 IS - 20 PB - Amer Soc Clinical Investigation Inc PY - 2020 SN - 2379-3708 ER - TY - JOUR AB - Immune checkpoint blockade has revolutionized cancer treatment, Patients developing immune mediated adverse events, such as colitis, appear to particularly benefit from immune checkpoint inhibition. Yet, the contributing mechanisms are largely unknown. We identified a systemic LP5 signature in melanoma patients with colitis following anti-cytotoxic T lymphocyte-associated antigen 4 (anti-CTLA-4) checkpoint inhibitor treatment and hypothesized that intestinal microbiota-derived LPS contributes to therapeutic efficacy. Because activation of immune cells within the tumor microenvironment is considered most promising to effectively control cancer, we analyzed human and murine melanoma for known sentinels of LPS. We identified mast cells (MCs) accumulating in and around melanomas and showed that effective melanoma immune control was dependent on LP5-activated MCs recruiting tumor-infiltrating effector T cells by secretion of CXCL10. Importantly, CXCL10 was also upregulated in human melanomas with immune regression and in patients with colitis induced by anti-CTLA-4 antibody. Furthermore, we demonstrate that CXCL10 upregulation and an MC signature at the site of melanomas are biomarkers for better patient survival, These findings provide conclusive evidence for a "Trojan horse treatment strategy" in which the plasticity of cancer-resident immune cells, such as MCs, is used as a target to boost tumor immune defense. AU - Kaesler, S.* AU - Wölbing, F.* AU - Kempf, W.E.* AU - Skabytska, Y. AU - Köberle, M.* AU - Volz, T.* AU - Sinnberg, T.* AU - Amaral, T.* AU - Möckel, S.* AU - Yazdi, A.* AU - Metzler, G.* AU - Schaller, M.* AU - Hartmann, K.* AU - Weide, B.* AU - Garbe, C.* AU - Rammensee, H.G.* AU - Röcken, M.* AU - Biedermann, T. C1 - 57012 C2 - 47432 CY - 2015 Manchester Rd, Ann Arbor, Mi 48104 Usa TI - Targeting tumor-resident mast cells for effective anti-melanoma immune responses. JO - JCI insight VL - 4 IS - 19 PB - Amer Soc Clinical Investigation Inc PY - 2019 SN - 2379-3708 ER - TY - JOUR AB - The increased formation of methylglyoxal (MG) under hyperglycemia is associated with the development of microvascular complications in patients with diabetes mellitus; however, the effects of elevated MG levels in vivo are poorly understood. In zebrafish, a transient knockdown of glyoxalase 1, the main MG detoxifying system, led to the elevation of endogenous MG levels and blood vessel alterations. To evaluate effects of a permanent knockout of glyoxalase 1 in vivo, glo1-/- zebrafish mutants were generated using CRISPR/Cas9. In addition, a diet-induced-obesity zebrafish model was used to analyze glo1-/- zebrafish under high nutrient intake. Glo1-/- zebrafish survived until adulthood without growth deficit and showed increased tissue MG concentrations. Impaired glucose tolerance developed in adult glo1-/- zebrafish and was indicated by increased postprandial blood glucose levels and postprandial S6 kinase activation. Challenged by an overfeeding period, fasting blood glucose levels in glo1-/- zebrafish were increased which translated into retinal blood vessel alterations. Thus, the data have identified a defective MG detoxification as a metabolic prerequisite and glyoxalase 1 alterations as a genetic susceptibility to the development of type 2 diabetes mellitus under high nutrition intake. AU - Lodd, E.* AU - Wiggenhauser, L.M.* AU - Morgenstern, J.* AU - Fleming, T.H.* AU - Poschet, G.* AU - Büttner, M.* AU - Tabler, C.T.* AU - Wohlfart, D.P.* AU - Nawroth, P.P. AU - Kroll, J.* C1 - 56767 C2 - 47382 TI - The combination of loss of glyoxalase1 and obesity results in hyperglycemia. JO - JCI insight VL - 4 IS - 12 PY - 2019 SN - 2379-3708 ER - TY - JOUR AB - Recent genetic examinations and multisteroid profiles have provided the basis for subclassification of aldosterone-producing adenomas (APAs). The objective of the current study was to produce a comprehensive, high-resolution mass spectrometry imaging (MSI) map of APAs in relation to morphometry, immunohistochemical profiles, mutational status, and clinical outcome. The study cohort comprised 136 patients with unilateral primary aldosteronism. Matrix-assisted laser desorption/ionization-Fourier transform-ion cyclotron resonance MSI was conducted, and metabolite profiles were analyzed with genotype/phenotype information, including digital image analysis from morphometry and IHC of steroidogenic enzymes. Distinct molecular signatures between KCNJ5- and CACNA1D-mutated APAs with significant differences of 137 metabolites, including metabolites of purine metabolism and steroidogenesis, were observed. Intratumor concentration of 18-oxocortisol and 18-hydroxycortisol were inversely correlated with the staining intensity of CYP11B1. Lower staining intensity of CYP11B1 and higher levels of 18-oxocortisol were associated with a higher probability of complete clinical success after surgery. The present study demonstrates distinct metabolomic profiles of APAs in relation to tumor genotype. In addition, we reveal an inverse correlation between cortisol derivatives and CYP11B1 and the impact of 18-oxocortisol and CYP11B1 on clinical outcome, which provides unprecedented insights into the pathophysiology, clinical features, and steroidogenesis of APAs. AU - Murakami, M.* AU - Rhayem, Y.* AU - Kunzke, T. AU - Sun, N. AU - Feuchtinger, A. AU - Ludwig, P.* AU - Strom, T.M. AU - Gomez-Sanchez, C.* AU - Knösel, T.* AU - Kirchner, T.* AU - Williams, T.A.* AU - Reincke, M.* AU - Walch, A.K. AU - Beuschlein, F.* C1 - 56854 C2 - 47304 TI - In situ metabolomics of aldosterone-producing adenomas. JO - JCI insight VL - 4 IS - 17 PY - 2019 SN - 2379-3708 ER - TY - JOUR AB - Lung transplantation (LTx) is the only therapeutic option for many patients with chronic lung disease. However, long-term survival after LTx is severely compromised by chronic rejection (chronic lung allograft dysfunction [CLAD]), which affects 50% of recipients after 5 years. The underlying mechanisms for CLAD are poorly understood, largely due to a lack of clinically relevant animal models, but lymphocytic bronchiolitis is an early sign of CLAD. Here, we report that lymphocytic bronchiolitis occurs early in a long-term murine orthotopic LTx model, based on a single mismatch (grafts from HLA-A2:B6–knockin donors transplanted into B6 recipients). Lymphocytic bronchiolitis is followed by formation of B cell–dependent lymphoid follicles that induce adjacent bronchial epithelial cell dysfunction in a spatiotemporal fashion. B cell deficiency using recipient μMT–/– mice prevented intrapulmonary lymphoid follicle formation and lymphocytic bronchiolitis. Importantly, selective inhibition of the follicle-organizing receptor EBI2, using genetic deletion or pharmacologic inhibition, prevented functional and histological deterioration of mismatched lung grafts. In sum, we provided what we believe to be a mouse model of chronic rejection and lymphocytic bronchiolitis after LTx and identified intrapulmonary lymphoid follicle formation as a target for pharmacological intervention of long-term allograft dysfunction after LTx. AU - Smirnova, N.F. AU - Conlon, T.M. AU - Morrone, C. AU - Dorfmuller, P.* AU - Humbert, M.* AU - Stathopoulos, G.T. AU - Umkehrer, S.* AU - Pfeiffer, F.* AU - Yildirim, A.Ö. AU - Eickelberg, O. C1 - 55444 C2 - 46358 CY - 2015 Manchester Rd, Ann Arbor, Mi 48104 Usa TI - Inhibition of B cell–dependent lymphoid follicle formation prevents lymphocytic bronchiolitis after lung transplantation. JO - JCI insight VL - 4 IS - 3 PB - Amer Soc Clinical Investigation Inc PY - 2019 SN - 2379-3708 ER - TY - JOUR AB - Chronic obstructive pulmonary disease (COPD) is a highly prevalent and devastating condition for which no curative treatment is available. Exaggerated lung cell senescence may be a major pathogenic factor. Here, we investigated the potential role for mTOR signaling in lung cell senescence and alterations in COPD using lung tissue and derived cultured cells from patients with COPD and from age- and sex-matched control smokers. Cell senescence in COPD was linked to mTOR activation, and mTOR inhibition by low-dose rapamycin prevented cell senescence and inhibited the proinflammatory senescence-associated secretory phenotype. To explore whether mTOR activation was a causal pathogenic factor, we developed transgenic mice exhibiting mTOR overactivity in lung vascular cells or alveolar epithelial cells. In this model, mTOR activation was sufficient to induce lung cell senescence and to mimic COPD lung alterations, with the rapid development of lung emphysema, pulmonary hypertension, and inflammation. These findings support a causal relationship between mTOR activation, lung cell senescence, and lung alterations in COPD, thereby identifying the mTOR pathway as a potentially new therapeutic target in COPD. AU - Houssaini, A. AU - Breau, M.* AU - Kebe, K.* AU - Abid, S.* AU - Marcos, E.* AU - Lipskaia, L.* AU - Rideau, D.* AU - Parpaleix, A.* AU - Huang, J.* AU - Amsellem, V.* AU - Vienney, N.* AU - Validire, P.* AU - Maitre, B.* AU - Attwe, A.* AU - Lukas, C. AU - Vindrieux, D.* AU - Boczkowski, J.* AU - Derumeaux, G.* AU - Pende, M.* AU - Bernard, D.* AU - Meiners, S. AU - Adnot, S.* C1 - 52931 C2 - 44438 TI - mTOR pathway activation drives lung cell senescence and emphysema. JO - JCI insight VL - 3 IS - 3 PY - 2018 SN - 2379-3708 ER - TY - JOUR AB - Autoimmune-mediated destruction of pancreatic islet β cells results in type 1 diabetes (T1D). Serum islet autoantibodies usually develop in genetically susceptible individuals in early childhood before T1D onset, with multiple islet autoantibodies predicting diabetes development. However, most at-risk children remain islet-antibody negative, and no test currently identifies those likely to seroconvert. We sought a genomic signature predicting seroconversion risk by integrating longitudinal peripheral blood gene expression profiles collected in high-risk children included in the BABYDIET and DIPP cohorts, of whom 50 seroconverted. Subjects were followed for 10 years to determine time of seroconversion. Any cohort effect and the time of seroconversion were corrected to uncover genes differentially expressed (DE) in seroconverting children. Gene expression signatures associated with seroconversion were evident during the first year of life, with 67 DE genes identified in seroconverting children relative to those remaining antibody negative. These genes contribute to T cell-, DC-, and B cell-related immune responses. Near-birth expression of ADCY9, PTCH1, MEX3B, IL15RA, ZNF714, TENM1, and PLEKHA5, along with HLA risk score predicted seroconversion (AUC 0.85). The ubiquitin-proteasome pathway linked DE genes and T1D susceptibility genes. Therefore, a gene expression signature in infancy predicts risk of seroconversion. Ubiquitination may play a mechanistic role in diabetes progression. AU - Mehdi, A.M.* AU - Hamilton-Williams, E.E.* AU - Cristino, A.* AU - Ziegler, A.-G. AU - Bonifacio, E.* AU - Le Cao, K.A.* AU - Harris, M.H.* AU - Thomas, R.* C1 - 53218 C2 - 44484 TI - A peripheral blood transcriptomic signature predicts autoantibody development in infants at risk of type 1 diabetes. JO - JCI insight VL - 3 IS - 5 PY - 2018 SN - 2379-3708 ER - TY - JOUR AB - Type 1 diabetes (T1D) is a chronic disease characterized by an autoimmune-mediated destruction of insulin-producing pancreatic β cells. Environmental factors such as viruses play an important role in the onset of T1D and interact with predisposing genes. Recent data suggest that viral infection of human islets leads to a decrease in insulin production rather than β cell death, suggesting loss of β cell identity. We undertook this study to examine whether viral infection could induce human β cell dedifferentiation. Using the functional human β cell line EndoC-βH1, we demonstrate that polyinosinic-polycytidylic acid (PolyI:C), a synthetic double-stranded RNA that mimics a byproduct of viral replication, induces a decrease in β cell-specific gene expression. In parallel with this loss, the expression of progenitor-like genes such as SOX9 was activated following PolyI:C treatment or enteroviral infection. SOX9 was induced by the NF-κB pathway and also in a paracrine non-cell-autonomous fashion through the secretion of IFN-α. Lastly, we identified SOX9 targets in human β cells as potentially new markers of dedifferentiation in T1D. These findings reveal that inflammatory signaling has clear implications in human β cell dedifferentiation. AU - Oshima, M.* AU - Knoch, K.-P. AU - Diedisheim, M.* AU - Petzold, A. AU - Cattan, P.* AU - Bugliani, M.* AU - Marchetti, P.* AU - Choudhary, P.* AU - Huang, G.C.* AU - Bornstein, S.R.* AU - Solimena, M. AU - Albagli-Curiel, O.* AU - Scharfmann, R.* C1 - 53086 C2 - 44327 TI - Virus-like infection induces human β cell dedifferentiation. JO - JCI insight VL - 3 IS - 3 PY - 2018 SN - 2379-3708 ER - TY - JOUR AB - Mesenchymal TNF signaling is etiopathogenic for inflammatory diseases such as rheumatoid arthritis and spondyloarthritis (SpA). The role of Tnfr1 in arthritis has been documented; however, Tnfr2 functions are unknown. Here, we investigate the mesenchymal-specific role of Tnfr2 in the Tnf(1ARE) mouse model of SpA in arthritis and heart valve stenosis comorbidity by cell-specific, Col6a1-cre-driven gene targeting. We find that TNF/Tnfr2 signaling in resident synovial fibroblasts (SFs) and valvular interstitial cells (VICs) is detrimental for both pathologies, pointing to common cellular mechanisms. In contrast, systemic Tnfr2 provides protective signaling, since its complete deletion leads to severe deterioration of both pathologies. SFs and VICs lacking Tnfr2 fail to acquire pathogenic activated phenotypes and display increased expression of antiinflammatory cytokines associated with decreased Akt signaling. Comparative RNA sequencing experiments showed that the majority of the deregulated pathways in Tnf(1ARE) mesenchymal-origin SFs and VICs, including proliferation, inflammation, migration, and disease-specific genes, are regulated by Tnfr2; thus, in its absence, they are maintained in a quiescent nonpathogenic state. Our data indicate a pleiotropy of Tnfr2 functions, with mesenchymal Tnfr2 driving cell activation and arthritis/valve stenosis pathogenesis only in the presence of systemic Tnfr2, whereas nonmesenchymal Tnfr2 overcomes this function, providing protective signals and, thus, containing both pathologies. AU - Sakkou, M.* AU - Chouvardas, P.* AU - Ntari, L.* AU - Prados, A.* AU - Moreth, K. AU - Fuchs, H. AU - Gailus-Durner, V. AU - Hrabě de Angelis, M. AU - Denis, M.C.* AU - Karagianni, N.* AU - Kollias, G.* C1 - 53369 C2 - 44544 CY - 2015 Manchester Rd, Ann Arbor, Mi 48104 Usa TI - Mesenchymal TNFR2 promotes the development of polyarthritis and comorbid heart valve stenosis. JO - JCI insight VL - 3 IS - 7 PB - Amer Soc Clinical Investigation Inc PY - 2018 SN - 2379-3708 ER - TY - JOUR AB - BACKGROUND: Adrenal aldosterone excess is the most common cause of secondary hypertension and is associated with increased cardiovascular morbidity. However, adverse metabolic risk in primary aldosteronism extends beyond hypertension, with increased rates of insulin resistance, type 2 diabetes, and osteoporosis, which cannot be easily explained by aldosterone excess. METHODS: We performed mass spectrometry-based analysis of a 24-hour urine steroid metabolome in 174 newly diagnosed patients with primary aldosteronism (103 unilateral adenomas, 71 bilateral adrenal hyperplasias) in comparison to 162 healthy controls, 56 patients with endocrine inactive adrenal adenoma, 104 patients with mild subclinical, and 47 with clinically overt adrenal cortisol excess. We also analyzed the expression of cortisol-producing CYP11B1 and aldosterone-producing CYP11B2 enzymes in adenoma tissue from 57 patients with aldosterone-producing adenoma, employing immunohistochemistry with digital image analysis. RESULTS: Primary aldosteronism patients had significantly increased cortisol and total glucocorticoid metabolite excretion (all P < 0.001), only exceeded by glucocorticoid output in patients with clinically overt adrenal Cushing syndrome. Several surrogate parameters of metabolic risk correlated significantly with glucocorticoid but not mineralocorticoid output. Intratumoral CYP11B1 expression was significantly associated with the corresponding in vivo glucocorticoid excretion. Unilateral adrenalectomy resolved both mineralocorticoid and glucocorticoid excess. Postoperative evidence of adrenal insufficiency was found in 13 (29%) of 45 consecutively tested patients. CONCLUSION: Our data indicate that glucocorticoid cosecretion is frequently found in primary aldosteronism and contributes to associated metabolic risk. Mineralocorticoid receptor antagonist therapy alone may not be sufficient to counteract adverse metabolic risk in medically treated patients with primary aldosteronism. FUNDING: Medical Research Council UK, Wellcome Trust, European Commission. AU - Arlt, W.* AU - Lang, K.* AU - Sitch, A.J.* AU - Dietz, A.S.* AU - Rhayem, Y.* AU - Bancos, I.* AU - Feuchtinger, A. AU - Chortis, V.* AU - Gilligan, L.C.* AU - Ludwig, P.* AU - Riester, A.* AU - Asbach, E.* AU - Hughes, B.A.* AU - O'Neil, D.M.* AU - Bidlingmaier, M.* AU - Tomlinson, J.W.* AU - Hassan-Smith, Z.K.* AU - Rees, D.A.* AU - Adolf, C.* AU - Hahner, S.* AU - Quinkler, M.* AU - Dekkers, T.* AU - Deinum, J.* AU - Biehl, M.* AU - Keevil, B.G.* AU - Shackleton, C.H.L.* AU - Deeks, J.J.* AU - Walch, A.K. AU - Beuschlein, F.* AU - Reincke, M.* C1 - 51006 C2 - 42623 TI - Steroid metabolome analysis reveals prevalent glucocorticoid excess in primary aldosteronism. JO - JCI insight VL - 2 IS - 8 PY - 2017 SN - 2379-3708 ER - TY - JOUR AB - BACKGROUND: Food intake is guided by homeostatic needs and by the reward value of food, yet the exact relation between the two remains unclear. The aim of this study was to investigate the influence of different metabolic states and hormonal satiety signaling on responses in neural reward networks. METHODS: Twenty-three healthy participants underwent functional magnetic resonance imaging while performing a task distinguishing between the anticipation and the receipt of either food- or monetary-related reward. Every participant was scanned twice in a counterbalanced fashion, both during a fasted state (after 24 hours fasting) and satiety. A functional connectivity analysis was performed to investigate the influence of satiety signaling on activation in neural reward networks. Blood samples were collected to assess hormonal satiety signaling. RESULTS: Fasting was associated with sensitization of the striatal reward system to the anticipation of food reward irrespective of reward magnitude. Furthermore, during satiety, individual ghrelin levels were associated with increased neural processing during the expectation of food-related reward. CONCLUSIONS: Our findings show that physiological hunger stimulates food consumption by specifically increasing neural processing during the expectation (i.e., incentive salience) but not the receipt of food-related reward. In addition, these findings suggest that ghrelin signaling influences hedonic-driven food intake by increasing neural reactivity during the expectation of food-related reward. These results provide insights into the neurobiological underpinnings of motivational processing and hedonic evaluation of food reward. TRIAL REGISTRATION: ClinicalTrials.gov NCT03081585. AU - Simon, J.J.* AU - Wetzel, A.* AU - Sinno, M.H.* AU - Skunde, M.* AU - Bendszus, M.* AU - Preissl, H. AU - Enck, P.* AU - Herzog, W.* AU - Friederich, H.C.* C1 - 51680 C2 - 43399 TI - Integration of homeostatic signaling and food reward processing in the human brain. JO - JCI insight VL - 2 IS - 15 PY - 2017 SN - 2379-3708 ER - TY - JOUR AB - Maladaptive epithelial repair from chronic injury is a common feature in fibrotic diseases, which in turn activates a pathogenic fibroblast response that produces excessive matrix deposition. Dysregulated microRNAs (miRs) can regulate expression of multiple genes and fundamentally alter cellular phenotypes during fibrosis. Although several miRs have been shown to be associated with lung fibrosis, the mechanisms by which miRs modulate epithelial behavior in lung fibrosis are lacking. Here, we identified miR-323a-3p to be downregulated in the epithelium of lungs with bronchiolitis obliterans syndrome (BOS) after lung transplantation, idiopathic pulmonary fibrosis (IPF), and murine bleomycin-induced fibrosis. Antagomirs for miR-323a-3p augment, and mimics suppress, murine lung fibrosis after bleomycin injury, indicating that this miR may govern profibrotic signals. We demonstrate that miR-323a-3p attenuates TGF-α and TGF-β signaling by directly targeting key adaptors in these important fibrogenic pathways. Moreover, miR-323a-3p lowers caspase-3 expression, thereby limiting programmed cell death from inducers of apoptosis and ER stress. Finally, we find that epithelial expression of miR-323a-3p modulates inhibitory crosstalk with fibroblasts. These studies demonstrate that miR-323a-3p has a central role in lung fibrosis that spans across murine and human disease, and downregulated expression by the lung epithelium releases inhibition of various profibrotic pathways to promote fibroproliferation. AU - Ge, L.* AU - Habiel, D.M.* AU - Hansbro, P.M.* AU - Kim, R.Y.* AU - Gharib, S.A.* AU - Edelman, J.D.* AU - Königshoff, M. AU - Parimon, T.* AU - Brauer, R.* AU - Huang, Y.* AU - Allen, J.* AU - Jiang, D.* AU - Kurkciyan, A.A.* AU - Mizuno, T.* AU - Stripp, B.R.* AU - Noble, P.W.* AU - Hogaboam, C.M.* AU - Chen, P.* C1 - 52947 C2 - 44252 TI - miR-323a-3p regulates lung fibrosis by targeting multiple profibrotic pathways. JO - JCI insight VL - 1 IS - 20 PY - 2016 SN - 2379-3708 ER -