TY - JOUR AB - SPNS1 is a lysosomal transporter mediating the salvage of lysoglycerophospholipids, the degradative products of lysosomal phospholipid catabolism. However, a role of lysolipid transport and salvage in regulating cellular lipid homeostasis and in disease is lacking. Here, we identified two families with biallelic SPNS1 loss-of-function variants that presented primarily with progressive liver and striated muscle injury. Patient fibroblasts accumulated lysophospholipids including lysoplasmalogens and cholesterol in lysosomes with reduced cellular plasmalogens. Notably, SPNS1 deficiency resulted in reduced biogenesis of cytosolic lipid droplets containing triglycerides and cholesteryl esters. Mechanistically, we found that lysophospholipids transported by SPNS1 into the cytosol quantitatively contributed to triglyceride synthesis while lysosomal buildup of lyso-ether-phospholipid inhibited lysosomal cholesterol egress, effects that were enhanced with inhibition of mTOR. These findings support a gene-disease association and reveal connectivity between lysosomal transport of lysophospholipids and storage of reserve cellular energy as triglyceride and in the regulation of cholesterol homeostasis, processes that become important under nutrient limitation. AU - He, M.* AU - Ding, M.* AU - Chocholoušková, M.* AU - Chin, C.F.* AU - Engvall, M.* AU - Malmgren, H.* AU - Wagner, M. AU - Lauffer, M.C.* AU - Heisinger, J.* AU - Malicdan, M.C.V.* AU - Allamand, V.* AU - Durbeej, M.* AU - Delgado-Vega, A.M.* AU - Sejersen, T.* AU - Nordgren, A.* AU - Torta, F.* AU - Silver, D.L.* C1 - 75063 C2 - 57785 TI - SPNS1 variants cause multi-organ disease and implicate lysophospholipid transport as critical for mTOR-regulated lipid homeostasis. JO - J. Clin. Invest. VL - 135 IS - 17 PY - 2025 SN - 0021-9738 ER - TY - JOUR AB - Fibrosis represents the uncontrolled replacement of parenchymal tissue with extracellular matrix (ECM) produced by myofibroblasts. While genetic fate-tracing and single-cell RNA-Seq technologies have helped elucidate fibroblast heterogeneity and ontogeny beyond fibroblast to myofibroblast differentiation, newly identified fibroblast populations remain ill defined, with respect to both the molecular cues driving their differentiation and their subsequent role in fibrosis. Using an unbiased approach, we identified the metalloprotease ADAMTS12 as a fibroblast-specific gene that is strongly upregulated during active fibrogenesis in humans and mice. Functional in vivo KO studies in mice confirmed that Adamts12 was critical during fibrogenesis in both heart and kidney. Mechanistically, using a combination of spatial transcriptomics and expression of catalytically active or inactive ADAMTS12, we demonstrated that the active protease of ADAMTS12 shaped ECM composition and cleaved hemicentin 1 (HMCN1) to enable the activation and migration of a distinct injury-responsive fibroblast subset defined by aberrant high JAK/STAT signaling. AU - Hoeft, K.* AU - Koch, L.* AU - Ziegler, S.* AU - Zhang, L.* AU - Luetke, S.* AU - Tanzer, M.C.* AU - Mohanta, D.* AU - Schumacher, D.* AU - Schreibing, F.* AU - Long, Q.* AU - Kim, H.* AU - Klinkhammer, B.M.* AU - Schikarski, C.* AU - Maryam, S.* AU - Baens, M.* AU - Hermann, J.* AU - Krieg, S.* AU - Peisker, F.* AU - De Laporte, L.* AU - Schaefer, G.J.L.* AU - Menzel, S.* AU - Jankowski, J.* AU - Humphreys, B.D.* AU - Wahida, A. AU - Schneider, R.K.* AU - Versele, M.* AU - Boor, P.* AU - Mann, M.* AU - Sengle, G.* AU - Hayat, S.* AU - Kramann, R.* C1 - 71731 C2 - 56400 CY - 2015 Manchester Rd, Ann Arbor, Mi 48104 Usa TI - ADAMTS12 promotes fibrosis by restructuring extracellular matrix to enable activation of injury-responsive fibroblasts. JO - J. Clin. Invest. VL - 134 IS - 18 PB - Amer Soc Clinical Investigation Inc PY - 2024 SN - 0021-9738 ER - TY - JOUR AB - Childhood neuroblastoma with MYCN-amplification is classified as high-risk and often relapses after intensive treatments. Immune checkpoint blockade therapy against the PD-1/L1 axis shows limited efficacy in neuroblastoma patients and the cancer intrinsic immune regulatory network is poorly understood. Here, we leverage genome-wide CRISPR/Cas9 screens and identify H2AFY as a resistance gene to the clinically approved PD-1 blocking antibody, nivolumab. Analysis of single-cell RNA sequencing datasets reveals that H2AFY mRNA is enriched in adrenergic cancer cells and is associated with worse patient survival. Genetic deletion of H2afy in MYCN-driven neuroblastoma cells reverts in vivo resistance to PD-1 blockade by eliciting activation of the adaptive and innate immunity. Mapping of the epigenetic and translational landscape demonstrates that H2afy deletion promotes cell transition to a mesenchymal-like state. With a multi-omics approach, we uncover H2AFY-associated genes that are functionally relevant and prognostic in patients. Altogether, our study elucidates the role of H2AFY as an epigenetic gatekeeper for cell states and immunogenicity in high-risk neuroblastoma. AU - Nagarajan, D.* AU - Parracho, R.T.* AU - Corujo, D.* AU - Xie, M.* AU - Kutkaite, G. AU - Olsen, T.K.* AU - Rúbies Bedós, M.* AU - Salehi, M.* AU - Baryawno, N.* AU - Menden, M.P. AU - Chen, X.* AU - Buschbeck, M.* AU - Mao, Y.* C1 - 71657 C2 - 56335 TI - Epigenetic regulation of cell state by H2AFY governs immunogenicity in high-risk neuroblastoma. JO - J. Clin. Invest. PY - 2024 SN - 0021-9738 ER - TY - JOUR AB - The β-secretase BACE1 is a central drug target for Alzheimer's disease. Clinically tested, BACE1-directed inhibitors also block the homologous protease BACE2. Yet, little is known about physiological BACE2 substrates and functions in vivo. Here, we identify BACE2 as the protease shedding the lymphangiogenic vascular endothelial growth factor receptor 3 (VEGFR3). Inactivation of BACE2, but not BACE1, inhibited shedding of VEGFR3 from primary human lymphatic endothelial cells (LECs) and reduced release of the shed, soluble VEGFR3 (sVEGFR3) ectodomain into the blood of mice, non-human primates and humans. Functionally, BACE2 inactivation increased full-length VEGFR3 and enhanced VEGFR3 signaling in LECs and also in vivo in zebrafish, where enhanced migration of LECs was observed. Thus, this study identifies BACE2 as a modulator of lymphangiogenic VEGFR3 signaling and demonstrates the utility of sVEGFR3 as a pharmacodynamic plasma marker for BACE2 activity in vivo, a prerequisite for developing BACE1-selective inhibitors for a safer prevention of Alzheimer's disease. AU - Schmidt, A.* AU - Hrupka, B.J.* AU - van Bebber, F.* AU - Sunil Kumar, S.* AU - Feng, X.* AU - Tschirner, S.K.* AU - Aßfalg, M.* AU - Müller, S.A.* AU - Hilger, L.S.* AU - Hofmann, L.I.* AU - Pigoni, M.* AU - Jocher, G.* AU - Voytyuk, I.* AU - Self, E.L.* AU - Ito, M.* AU - Hyakkoku, K.* AU - Yoshimura, A.* AU - Horiguchi, N.* AU - Feederle, R. AU - de Strooper, B.* AU - Schulte-Merker, S.* AU - Lammert, E.* AU - Moechars, D.* AU - Schmid, B.* AU - Lichtenthaler, S.F.* C1 - 70870 C2 - 55776 CY - 2015 Manchester Rd, Ann Arbor, Mi 48104 Usa TI - The Alzheimer's disease-linked protease BACE2 cleaves VEGFR3 and modulates its signaling. JO - J. Clin. Invest. VL - 134 IS - 16 PB - Amer Soc Clinical Investigation Inc PY - 2024 SN - 0021-9738 ER - TY - JOUR AB - Regulated exocytosis is initiated by increased Ca2+ concentrations in close spatial proximity to secretory granules, which is effectively prevented when the cell is at rest. Here we showed that exocytosis of zymogen granules in acinar cells was driven by Ca2+ directly released from acidic Ca2+ stores including secretory granules through NAADP-activated two-pore channels (TPCs). We identified OCaR1 (encoded by Tmem63a) as an organellar Ca2+ regulator protein integral to the membrane of secretory granules that controlled Ca2+ release via inhibition of TPC1 and TPC2 currents. Deletion of OCaR1 led to extensive Ca2+ release from NAADP-responsive granules under basal conditions as well as upon stimulation of GPCR receptors. Moreover, OCaR1 deletion exacerbated the disease phenotype in murine models of severe and chronic pancreatitis. Our findings showed OCaR1 as a gatekeeper of Ca2+ release that endows NAADP-sensitive secretory granules with an autoregulatory mechanism preventing uncontrolled exocytosis and pancreatic tissue damage. AU - Tsvilovskyy, V.* AU - Ottenheijm, R.* AU - Kriebs, U.* AU - Schütz, A.* AU - Diakopoulos, K.N.* AU - Jha, A.N.* AU - Bildl, W.* AU - Wirth, A.* AU - Böck, J.* AU - Jaślan, D.* AU - Ferro, I.* AU - Taberner, F.J.* AU - Kalinina, O.* AU - Hildebrand, S.* AU - Wissenbach, U.* AU - Weissgerber, P.* AU - Vogt, D.* AU - Eberhagen, C. AU - Mannebach, S.* AU - Berlin, M.* AU - Kuryshev, V.* AU - Schumacher, D.* AU - Philippaert, K.* AU - Camacho-Londoño, J.E.* AU - Mathar, I.* AU - Dieterich, C.* AU - Klugbauer, N.* AU - Biel, M.* AU - Wahl-Schott, C.* AU - Lipp, P.* AU - Flockerzi, V.* AU - Zischka, H. AU - Algül, H.* AU - Lechner, S.G.* AU - Lesina, M.* AU - Grimm, C.* AU - Fakler, B.* AU - Schulte, U.* AU - Muallem, S.* AU - Freichel, M.* C1 - 70363 C2 - 55531 CY - 2015 Manchester Rd, Ann Arbor, Mi 48104 Usa TI - OCaR1 endows exocytic vesicles with autoregulatory competence by preventing uncontrolled Ca2+ release, exocytosis, and pancreatic tissue damage. JO - J. Clin. Invest. VL - 134 IS - 7 PB - Amer Soc Clinical Investigation Inc PY - 2024 SN - 0021-9738 ER - TY - JOUR AU - Bass, J.* AU - Tschöp, M.H. AU - Beutler, L.R.* C1 - 67330 C2 - 54173 CY - 2015 Manchester Rd, Ann Arbor, Mi 48104 Usa TI - Dual gut hormone receptor agonists for diabetes and obesity. JO - J. Clin. Invest. VL - 133 IS - 3 PB - Amer Soc Clinical Investigation Inc PY - 2023 SN - 0021-9738 ER - TY - JOUR AB - Cell senescence (CS) is at the nexus between aging and associated chronic disorders, and aging increases the burden of CS in all major metabolic tissues. However, CS is also increased in adult obesity, type 2 diabetes (T2D), and nonalcoholic fatty liver disease independent of aging. Senescent tissues are characterized by dysfunctional cells and increased inflammation, and both progenitor cells and mature, fully differentiated and nonproliferating cells are afflicted. Recent studies have shown that hyperinsulinemia and associated insulin resistance (IR) promote CS in both human adipose and liver cells. Similarly, increased CS promotes cellular IR, showing their interdependence. Furthermore, the increased adipose CS in T2D is independent of age, BMI, and degree of hyperinsulinemia, suggesting premature aging. These results suggest that senomorphic/senolytic therapy may become important for treating these common metabolic disorders. AU - Spinelli, R.* AU - Baboota, R.K.* AU - Gogg, S.* AU - Beguinot, F.* AU - Blüher, M. AU - Nerstedt, A.* AU - Smith, U.* C1 - 69429 C2 - 53861 CY - 2015 Manchester Rd, Ann Arbor, Mi 48104 Usa TI - Increased cell senescence in human metabolic disorders. JO - J. Clin. Invest. VL - 133 IS - 12 PB - Amer Soc Clinical Investigation Inc PY - 2023 SN - 0021-9738 ER - TY - JOUR AB - Activated SUMOylation is a hallmark of cancer. Starting from a targeted screening for SUMO-regulated immune evasion mechanisms, we identified an evolutionarily conserved function of activated SUMOylation, which attenuated the immunogenicity of tumor cells. Activated SUMOylation allowed cancer cells to evade CD8+ T cell-mediated immunosurveillance by suppressing the MHC class I (MHC-I) antigen-processing and presentation machinery (APM). Loss of the MHC-I APM is a frequent cause of resistance to cancer immunotherapies, and the pharmacological inhibition of SUMOylation (SUMOi) resulted in reduced activity of the transcriptional repressor scaffold attachment factor B (SAFB) and induction of the MHC-I APM. Consequently, SUMOi enhanced the presentation of antigens and the susceptibility of tumor cells to CD8+ T cell-mediated killing. Importantly, SUMOi also triggered the activation of CD8+ T cells and thereby drove a feed-forward loop amplifying the specific antitumor immune response. In summary, we showed that activated SUMOylation allowed tumor cells to evade antitumor immunosurveillance, and we have expanded the understanding of SUMOi as a rational therapeutic strategy for enhancing the efficacy of cancer immunotherapies. AU - Demel, U.M.* AU - Boger, M.* AU - Yousefian, S.* AU - Grunert, C.* AU - Zhang, L.* AU - Hotz, P.W.* AU - Gottschlich, A.* AU - Köse, H.* AU - Isaakidis, K.* AU - Vonficht, D.* AU - Grünschläger, F.* AU - Rohleder, E.* AU - Wagner, K.* AU - Dönig, J.* AU - Igl, V.* AU - Brzezicha, B.* AU - Baumgartner, F.* AU - Habringer, S.* AU - Löber, J.* AU - Chapuy, B.* AU - Weidinger, C.* AU - Kobold, S. AU - Busse, A.B.* AU - Müller, S.* AU - Wirth, M.* AU - Schick, M.* AU - Keller, U.* C1 - 65031 C2 - 52144 TI - Activated SUMOylation restricts MHC class I antigen presentation to confer immune evasion in cancer. JO - J. Clin. Invest. VL - 132 IS - 9 PY - 2022 SN - 0021-9738 ER - TY - JOUR AB - The etiology of type 1 diabetes has polygenic and environmental determinants that lead to autoimmune responses against pancreatic β cells and promote β cell death. The autoimmunity is considered silent without metabolic consequences until late preclinical stages,and it remains unknown how early in the disease process the pancreatic β cell is compromised. To address this, we investigated preprandial nonfasting and postprandial blood glucose concentrations and islet autoantibody development in 1,050 children with high genetic risk of type 1 diabetes. Pre- and postprandial blood glucose decreased between 4 and 18 months of age and gradually increased until the final measurements at 3.6 years of age. Determinants of blood glucose trajectories in the first year of life included sex, body mass index, glucose-related genetic risk scores, and the type 1 diabetes-susceptible INS gene. Children who developed islet autoantibodies had early elevations in blood glucose concentrations. A sharp and sustained rise in postprandial blood glucose was observed at around 2 months prior to autoantibody seroconversion, with further increases in postprandial and, subsequently, preprandial values after seroconversion. These findings show heterogeneity in blood glucose control in infancy and early childhood and suggest that islet autoimmunity is concurrent or subsequent to insults on the pancreatic islets. AU - Warncke, K. AU - Weiss, A. AU - Achenbach, P. AU - von dem Berge, T.* AU - Berner, R.* AU - Casteels, K.* AU - Groele, L.* AU - Hatzikotoulas, K. AU - Hommel, A. AU - Kordonouri, O.* AU - Elding Larsson, H.* AU - Lundgren, M.* AU - Marcus, B.A. AU - Snape, M.D.* AU - Szypowska, A.* AU - Todd, J.A.* AU - Bonifacio, E. AU - Ziegler, A.-G. C1 - 66458 C2 - 52817 TI - Elevations in blood glucose before and after the appearance of islet autoantibodies in children. JO - J. Clin. Invest. VL - 132 IS - 20 PY - 2022 SN - 0021-9738 ER - TY - JOUR AB - Cortical spreading depression (CSD), a wave of depolarization followed by depression of cortical activity, is a pathophysiological process implicated in migraine with aura and various other brain pathologies, such as ischemic stroke and traumatic brain injury. To gain insight into the pathophysiology of CSD, we generated a mouse model for a severe monogenic subtype of migraine with aura, familial hemiplegic migraine type 3 (FHM3). FHM3 is caused by mutations in SCN1A, encoding the voltage-gated Na+ channel NaV1.1 predominantly expressed in inhibitory interneurons. Homozygous Scn1aL1649Q knock-in mice died prematurely, whereas heterozygous mice had a normal lifespan. Heterozygous Scn1aL1649Q knock-in mice compared to wildtype mice displayed a significantly enhanced susceptibility to CSD. We found L1649Q to cause a gain-of-function effect with an impaired Na+-channel inactivation and increased ramp Na+-currents leading to hyperactivity of fast-spiking inhibitory interneurons. Brain slice recordings using K+-sensitive electrodes revealed an increase in extracellular K+ in the early phase of CSD in heterozygous mice, likely representing the mechanistic link between interneuron hyperactivity and CSD initiation. The neuronal phenotype and premature death of homozygous Scn1aL1649Q knock-in mice was partially rescued by GS967, a blocker of persistent Na+-currents. Collectively, our findings identify interneuron hyperactivity as a mechanism to trigger CSD. AU - Auffenberg, E.* AU - Hedrich, U.B.S.* AU - Barbieri, R.* AU - Miely, D.* AU - Groschup, B.* AU - Wuttke, T.V.* AU - Vogel, N.* AU - Lührs, P.* AU - Zanardi, I.* AU - Bertelli, S.* AU - Spielmann, N. AU - Gailus-Durner, V. AU - Fuchs, H. AU - Hrabě de Angelis, M. AU - Pusch, M.C.* AU - Dichgans, M.* AU - Lerche, H.* AU - Gavazzo, P.* AU - Plesnila, N.* AU - Freilinger, T.* C1 - 63029 C2 - 51110 CY - 2015 Manchester Rd, Ann Arbor, Mi 48104 Usa TI - Hyperexcitable interneurons trigger cortical spreading depression in an Scn1a migraine model. JO - J. Clin. Invest. VL - 131 IS - 21 PB - Amer Soc Clinical Investigation Inc PY - 2021 SN - 0021-9738 ER - TY - JOUR AB - Genetic factors undoubtedly affect the development of congenital heart disease (CHD) but still remain ill defined. We sought to identify genetic risk factors associated with CHD and to accomplish a functional analysis of SNP-carrying genes. We performed a genome-wide association study (GWAS) of 4034 White patients with CHD and 8486 healthy controls. One SNP on chromosome 5q22.2 reached genome-wide significance across all CHD phenotypes and was also indicative for septal defects. One region on chromosome 20p12.1 pointing to the MACROD2 locus identified 4 highly significant SNPs in patients with transposition of the great arteries (TGA). Three highly significant risk variants on chromosome 17q21.32 within the GOSR2 locus were detected in patients with anomalies of thoracic arteries and veins (ATAV). Genetic variants associated with ATAV are suggested to influence the expression of WNT3, and the variant rs870142 related to septal defects is proposed to influence the expression of MSX1. We analyzed the expression of all 4 genes during cardiac differentiation of human and murine induced pluripotent stem cells in vitro and by single-cell RNA-Seq analyses of developing murine and human hearts. Our data show that MACROD2, GOSR2, WNT3, and MSX1 play an essential functional role in heart development at the embryonic and newborn stages. AU - Lahm, H.* AU - Jia, M.* AU - Dreßen, M.* AU - Wirth, F.* AU - Puluca, N.* AU - Gilsbach, R.* AU - Keavney, B.D.* AU - Cleuziou, J.* AU - Beck, N.* AU - Bondareva, O.* AU - Dzilic, E.* AU - Burri, M.* AU - König, K.C.* AU - Ziegelmüller, J.A.* AU - Abou-Ajram, C.* AU - Neb, I.* AU - Zhang, Z.* AU - Doppler, S.A.* AU - Mastantuono, E. AU - Lichtner, P. AU - Eckstein, G.N. AU - Hörer, J.* AU - Ewert, P.* AU - Priest, J.R.* AU - Hein, L.* AU - Lange, R.* AU - Meitinger, T. AU - Cordell, H.J.* AU - Müller-Myhsok, B.* AU - Krane, M.* C1 - 61153 C2 - 49775 CY - 2015 Manchester Rd, Ann Arbor, Mi 48104 Usa TI - Congenital heart disease risk loci identified by genome-wide association study in European patients. JO - J. Clin. Invest. VL - 131 IS - 2 PB - Amer Soc Clinical Investigation Inc PY - 2021 SN - 0021-9738 ER - TY - JOUR AB - Vascular stability and tone are maintained by contractile smooth muscle cells (VSMCs). However, injury-induced growth factors stimulate a contractile-synthetic phenotypic modulation which increases susceptibility to abdominal aortic aneurysm (AAA). As a regulator of embryonic VSMC differentiation, we hypothesized that Thymosin beta 4 (T beta 4) may function to maintain healthy vasculature throughout postnatal life. This was supported by the identification of an interaction with low density lipoprotein receptor related protein 1 (LRP1), an endocytic regulator of platelet-derived growth factor BB (PDGF-BB) signaling and VSMC proliferation. LRP1 variants have been implicated by genome-wide association studies with risk of AAA and other arterial diseases. T beta 4-null mice displayed aortic VSMC and elastin defects that phenocopy those of LRP1 mutants, and their compromised vascular integrity predisposed them to Angiotensin II-induced aneurysm formation. Aneurysmal vessels were characterized by enhanced VSMC phenotypic modulation and augmented PDGFR-beta signaling. In vitro, enhanced sensitivity to PDGF-BB upon loss of T beta 4 was associated with dysregulated endocytosis, with increased recycling and reduced lysosomal targeting of LRP1-PDGFR-beta. Accordingly, the exacerbated aneurysmal phenotype in T beta 4-null mice was rescued upon treatment with the PDGFR-beta antagonist Imatinib. Our study identifies T beta 4 as a key regulator of LRP1 for maintaining vascular health, and provides insights into the mechanisms of growth factor-controlled VSMC phenotypic modulation underlying aortic disease progression. AU - Munshaw, S.* AU - Bruche, S.* AU - Redpath, A.N.* AU - Jones, A. AU - Patel, J.* AU - Dube, K.N.* AU - Lee, R.* AU - Hester, S.S.* AU - Davies, R.* AU - Neal, G.* AU - Handa, A.* AU - Sattler, M. AU - Fischer, R.* AU - Channon, K.M.* AU - Smart, N.* C1 - 62123 C2 - 50623 CY - 2015 Manchester Rd, Ann Arbor, Mi 48104 Usa TI - Thymosin β4 protects against aortic aneurysm via endocytic regulation of growth factor signaling. JO - J. Clin. Invest. VL - 131 IS - 10 PB - Amer Soc Clinical Investigation Inc PY - 2021 SN - 0021-9738 ER - TY - JOUR AB - BACKGROUND: There is considerable variability in COVID-19 outcomes amongst younger adults-and some of this variation may be due to genetic predisposition. METHODS: We combined individual level data from 13,888 COVID-19 patients (N=7,185 hospitalized) from 17 cohorts in nine countries to assess the association of the major common COVID-19 genetic risk factor (chromosome 3 locus tagged by rs10490770) with mortality, COVID-19-related complications and laboratory values. We next performed meta-analyses using FinnGen and the Columbia University COVID-19 Biobank. RESULTS: We found that rs10490770 risk allele carriers experienced an increased risk of all-cause mortality (HR 1.4, 95%CI 1.2-1.7). Risk allele carriers had increased odds of several COVID-19 complications: severe respiratory failure (OR 2.1, 95%CI 1.6-2.6), venous thromboembolism (OR 1.7, 95%CI 1.2-2.4), and hepatic injury (OR 1.5, 95%CI 1.2-2.0). Risk allele carriers ≤60 years had higher odds of death or severe respiratory failure (OR 2.7, 95%CI 1.8-3.9) compared to those >60 years (OR 1.5, 95%CI 1.2-1.8, interaction-p=0.038). Amongst individuals ≤60 years who died or experienced severe respiratory failure, 32.3% were risk variant carriers, compared to 13.9% of those not experiencing these outcomes. The genetic risk improved the prediction of death or severe respiratory failure similarly to, or better than, most established clinical risk factors. CONCLUSIONS: The major common COVID-19 genetic risk factor is associated with increased risks of morbidity and mortality, which are more pronounced amongst individuals ≤60 years. The effect was similar in magnitude and more common than most established clinical risk factors, suggesting potential implications for future clinical risk management. AU - Nakanishi, T.* AU - Pigazzini, S.* AU - Degenhardt, F.* AU - Cordioli, M.* AU - Butler-Laporte, G.* AU - Maya-Miles, D.* AU - Bujanda, L.* AU - Bouysran, Y.* AU - Niemi, M.E.* AU - Palom, A.* AU - Ellinghaus, D.* AU - Khan, A.* AU - Martínez-Bueno, M.* AU - Rolker, S.* AU - Amitrano, S.* AU - Roade Tato, L.* AU - Fava, F.* AU - Spinner, C.D.* AU - Prati, D.* AU - Bernardo, D.* AU - Garcia, F.* AU - Darcis, G.* AU - Fernandez-Cadenas, I.* AU - Holter, J.C.* AU - Banales, J.M.* AU - Frithiof, R.* AU - Kiryluk, K.* AU - Duga, S.* AU - Asselta, R.* AU - Pereira, A.C.* AU - Romero-Gómez, M.* AU - Nafría-Jiménez, B.* AU - Hov, J.R.* AU - Migeotte, I.* AU - Renieri, A.* AU - Planas, A.M.* AU - Ludwig, K.U.* AU - Buti, M.* AU - Rahmouni, S.* AU - Alarcón-Riquelme, M.E.* AU - Schulte, E.C. AU - Franke, A.* AU - Karlsen, T.H.* AU - Valenti, L.* AU - Zeberg, H.* AU - Richards, J.B.* AU - Ganna, A.* C1 - 63183 C2 - 51381 CY - 2015 Manchester Rd, Ann Arbor, Mi 48104 Usa TI - Age-dependent impact of the major common genetic risk factor for COVID-19 on severity and mortality. JO - J. Clin. Invest. VL - 131 IS - 23 PB - Amer Soc Clinical Investigation Inc PY - 2021 SN - 0021-9738 ER - TY - JOUR AB - Leber's hereditary optic neuropathy (LHON) is the most frequent mitochondrial disease and was the first to be genetically defined by a point mutation in the mitochondrial DNA (mtDNA). A molecular diagnosis is reached in up to 95%, the vast majority of which are accounted for by three mutations within mitochondrial complex I (CI) subunit encoding genes in the mtDNA (mtLHON). Here, we resolve the enigma of LHON in the absence of pathogenic mtDNA mutations. We describe biallelic mutations in a nuclear encoded gene, DNAJC30, in 33 unsolved patients from 29 families and establish an autosomal recessive mode of inheritance for LHON (arLHON), which to date has been a prime example of a maternally inherited disorder. Remarkably, all hallmarks of mtLHON are recapitulated, including incomplete penetrance, male predominance, and significant idebenone responsivity. Moreover, by tracking protein turnover in patient-derived cell lines and a DNAJC30-knock-out cellular model, we measure reduced turnover of specific CI N-module subunits and a resultant impairment of CI function. This demonstrates DNAJC30 is to be a chaperone protein needed for the efficient exchange of CI subunits exposed to reactive oxygen species and integral to a mitochondrial CI repair mechanism, thereby providing the first example of a disease resulting from impaired exchange of assembled respiratory chain subunits. AU - Stenton, S. AU - Sheremet, N.L.* AU - Catarino, C.B.* AU - Andreeva, N.* AU - Assouline, Z.* AU - Barboni, P.* AU - Barel, O.* AU - Berutti, R. AU - Bychkov, I.O.* AU - Caporali, L.* AU - Capristo, M.* AU - Carbonelli, M.* AU - Cascavilla, M.L.* AU - Charbel Issa, P.* AU - Freisinger, P.* AU - Gerber, S.* AU - Ghezzi, D.* AU - Graf, E. AU - Heidler, J.* AU - Hempel, M.* AU - Héon, E.* AU - Itkis, Y.S.* AU - Javasky, E.* AU - Kaplan, J.* AU - Kopajtich, R. AU - Kornblum, C.* AU - Kovács-Nagy, R.* AU - Krylova, T.* AU - Kunz, W.S.* AU - La Morgia, C.* AU - Lamperti, C.* AU - Ludwig, C.* AU - Malacarne, P.F.* AU - Maresca, A.* AU - Mayr, J.A.* AU - Meisterknecht, J.* AU - Nevinitsyna, T.* AU - Palombo, F.* AU - Pode-Shakked, B.* AU - Shmelkova, M.S.* AU - Strom, T.M.* AU - Tagliavini, F.* AU - Tzadok, M.* AU - van der Ven, A.T.* AU - Vignal-Clermont, C.* AU - Wagner, M. AU - Zakharova, E.* AU - Zhorzholadze, N.* AU - Rozet, J.M.* AU - Carelli, V.* AU - Tsygankova, P.* AU - Klopstock, T.* AU - Wittig, I.* AU - Prokisch, H. C1 - 61088 C2 - 49685 CY - 2015 Manchester Rd, Ann Arbor, Mi 48104 Usa TI - Impaired complex I repair causes recessive Leber's hereditary optic neuropathy. JO - J. Clin. Invest. VL - 131 IS - 6 PB - Amer Soc Clinical Investigation Inc PY - 2021 SN - 0021-9738 ER - TY - JOUR AB - Inherited optic neuropathies include complex phenotypes, mostly driven by mitochondrial dysfunction. We report an optic atrophy spectrum disorder, including retinal macular dystrophy and kidney insufficiency leading to transplantation, associated with mitochondrial DNA (mtDNA) depletion without accumulation of multiple deletions. By whole-exome sequencing, we identified mutations affecting the mitochondrial single-strand binding protein (SSBP1) in 4 families with dominant and 1 with recessive inheritance. We show that SSBP1 mutations in patient-derived fibroblasts variably affect the amount of SSBP1 protein and alter multimer formation, but not the binding to ssDNA. SSBP1 mutations impaired mtDNA, nucleoids, and 7S-DNA amounts as well as mtDNA replication, affecting replisome machinery. The variable mtDNA depletion in cells was reflected in severity of mitochondrial dysfunction, including respiratory efficiency, OXPHOS subunits, and complex amount and assembly. mtDNA depletion and cytochrome c oxidase-negative cells were found ex vivo in biopsies of affected tissues, such as kidney and skeletal muscle. Reduced efficiency of mtDNA replication was also reproduced in vitro, confirming the pathogenic mechanism. Furthermore, ssbp1 suppression in zebrafish induced signs of nephropathy and reduced optic nerve size, the latter phenotype complemented by WT mRNA but not by SSBP1 mutant transcripts. This previously unrecognized disease of mtDNA maintenance implicates SSBP1 mutations as a cause of human pathology. AU - Del Dotto, V.* AU - Ullah, F.* AU - Di Meo, I.* AU - Magini, P.* AU - Gusic, M. AU - Maresca, A.* AU - Caporali, L.* AU - Palombo, F.* AU - Tagliavini, F.* AU - Baugh, E.H.* AU - Macao, B.* AU - Szilagyi, Z.* AU - Peron, C.* AU - Gustafson, M.A.* AU - Khan, K.* AU - La Morgia, C.* AU - Barboni, P.* AU - Carbonelli, M.* AU - Valentino, M.L.* AU - Liguori, R.* AU - Shashi, V.* AU - Sullivan, J.* AU - Nagaraj, S.* AU - El-Dairi, M.* AU - Iannaccone, A.* AU - Cutcutache, I.* AU - Bertini, E.* AU - Carrozzo, R.* AU - Emma, F.* AU - Diomedi-Camassei, F.* AU - Zanna, C.* AU - Armstrong, M.* AU - Page, M.* AU - Stong, N.* AU - Boesch, S.* AU - Kopajtich, R. AU - Wortmann, S.B. AU - Sperl, W.* AU - Davis, E.E.* AU - Copeland, W.C.* AU - Seri, M.* AU - Falkenberg, M.* AU - Prokisch, H. AU - Katsanis, N.* AU - Tiranti, V.* AU - Pippucci, T.* AU - Carelli, V.* C1 - 57804 C2 - 47975 CY - 2015 Manchester Rd, Ann Arbor, Mi 48104 Usa SP - 108-125 TI - SSBP1 mutations cause mtDNA depletion underlying a complex optic atrophy disorder. JO - J. Clin. Invest. VL - 130 IS - 1 PB - Amer Soc Clinical Investigation Inc PY - 2020 SN - 0021-9738 ER - TY - JOUR AB - Recent genome-wide association studies (GWAS) identified DUSP8, encoding a dual-specificity phosphatase targeting mitogen-activated protein kinases, as a type 2 diabetes (T2D) risk gene. Here, we reveal that Dusp8 is a gatekeeper in the hypothalamic control of glucose homeostasis in mice and humans. Male, but not female, Dusp8 loss-of-function mice, either with global or corticotropin-releasing hormone neuron-specific deletion, had impaired systemic glucose tolerance and insulin sensitivity when exposed to high-fat diet (HFD). Mechanistically, we found impaired hypothalamic-pituitary-adrenal axis feedback, blunted sympathetic responsiveness, and chronically elevated corticosterone levels driven by hypothalamic hyperactivation of Jnk signaling. Accordingly, global/Jnk1 ablation, AAV-mediated Dusp8 overexpression in the mediobasal hypothalamus, or metyrapone-induced chemical adrenalectomy rescued the impaired glucose homeostasis of obese male Dusp8-KO mice, respectively. The sex-specific role of murine Dusp8 in governing hypothalamic Jnk signaling, insulin sensitivity, and systemic glucose tolerance was consistent with functional MRI data in human volunteers that revealed an association of the DUSP8 rs2334499 risk variant with hypothalamic insulin resistance in men. Further, expression of DUSP8 was increased in the infundibular nucleus of T2D humans. In summary, our findings suggest the GWAS-identified gene Dusp8 as a novel hypothalamic factor that plays a functional role in the etiology of T2D. AU - Schriever, S.C. AU - Kabra, D.G. AU - Pfuhlmann, K. AU - Baumann, P. AU - Baumgart, E.V. AU - Nagler, J. AU - Seebacher, F. AU - Harrison, L. AU - Irmler, M. AU - Kullmann, S. AU - Corrêa-da-Silva, F.* AU - Giesert, F. AU - Jain, R.* AU - Schug, H.* AU - Castel, J.* AU - Martinez, S.* AU - Wu, M. AU - Häring, H.-U. AU - Hrabě de Angelis, M. AU - Beckers, J. AU - Müller, T.D. AU - Stemmer, K. AU - Wurst, W. AU - Rozman, J. AU - Nogueiras, R.* AU - de Angelis, M. AU - Molkentin, J.D.* AU - Krahmer, N. AU - Yi, C.-X.* AU - Schmidt, M.V.* AU - Luquet, S.* AU - Heni, M. AU - Tschöp, M.H. AU - Pfluger, P.T. C1 - 59952 C2 - 48944 CY - 2015 Manchester Rd, Ann Arbor, Mi 48104 Usa SP - 6093-6108 TI - Type 2 diabetes risk gene Dusp8 regulates hypothalamic Jnk signaling and insulin sensitivity. JO - J. Clin. Invest. VL - 130 IS - 11 PB - Amer Soc Clinical Investigation Inc PY - 2020 SN - 0021-9738 ER - TY - JOUR AB - BACKGROUND. Given the heightened tolerance to self-starvation in anorexia nervosa (AN), a hypothalamic dysregulation of energy and glucose homeostasis has been hypothesized. Therefore, we investigated whether hypothalamic reactivity to glucose metabolism is impaired in AN.METHODS. Twenty-four participants with AN, 28 normal-weight participants, and 24 healthy participants with obesity underwent 2 MRI sessions in a single-blind, randomized, case-controlled crossover study. We used an intragastric infusion of glucose and water to bypass the cephalic phase of food intake. The responsivity of the hypothalamus and the crosstalk of the hypothalamus with reward-related brain regions were investigated using high-resolution MRI.RESULTS. Normal-weight control participants displayed the expected glucose-induced deactivation of hypothalamic activation, whereas patients with AN and participants with obesity showed blunted hypothalamic reactivity. Furthermore, patients with AN displayed blunted reactivity in the nucleus accumbens and amygdala. Compared with the normal-weight participants and control participants with obesity, the patients with AN failed to show functional connectivity between the hypothalamus and the reward-related brain regions during water infusion relative to glucose infusion. Finally, the patients with AN displayed typical baseline levels of peripheral appetite hormones during a negative energy balance.CONCLUSION. These results indicate that blunted hypothalamic glucose reactivity might be related to the pathophysiology of AN. This study provides insights for future research, as it is an extended perspective of the traditional primary nonhomeostatic understanding of the disease. AU - Simon, J.J.* AU - Stopyra, M.A.* AU - Mönning, E.* AU - Sailer, S.* AU - Lavandier, N.* AU - Kihm, L.P.* AU - Bendszus, M.* AU - Preissl, H. AU - Herzog, W.* AU - Friederich, H.C. C1 - 59510 C2 - 48887 CY - 2015 Manchester Rd, Ann Arbor, Mi 48104 Usa SP - 4094-4103 TI - Neuroimaging of hypothalamic mechanisms related to glucose metabolism in anorexia nervosa and obesity. JO - J. Clin. Invest. VL - 130 IS - 8 PB - Amer Soc Clinical Investigation Inc PY - 2020 SN - 0021-9738 ER - TY - JOUR AB - lschemic stroke is a predominant cause of disability worldwide, with thrombolytic or mechanical removal of the occlusion being the only therapeutic option. Reperfusion bears the risk of an acute deleterious calcium-dependent breakdown of the blood-brain barrier. Its mechanism, however, is unknown. Here, we identified type 5 NADPH oxidase (NOX5), a calciumactivated, ROS-forming enzyme, as the missing link. Using a humanized knockin (KI) mouse model and in vitro organotypic cultures, we found that reoxygenation or calcium overload increased brain ROS levels in a NOX5-dependent manner. In vivo, postischemic ROS formation, infarct volume, and functional outcomes were worsened in NOXS-KI mice. Of clinical and therapeutic relevance, in a human blood-barrier model, pharmacological NOX inhibition also prevented acute reoxygenationinduced leakage. Our data support further evaluation of poststroke recanalization in the presence of NOX inhibition for limiting stroke-induced damage. AU - Casas, A.I.* AU - Kleikers, P.W.M.* AU - Geuss, E.* AU - Langhauser, F.* AU - Adler, T. AU - Busch, D.H.* AU - Gailus-Durner, V. AU - Hrabě de Angelis, M. AU - Egea, J.* AU - Lopez, M.G.* AU - Kleinschnitz, C.* AU - Schmidt, H.H.H.W.* C1 - 55704 C2 - 46461 CY - 2015 Manchester Rd, Ann Arbor, Mi 48104 Usa SP - 1772-1778 TI - Calcium-dependent blood-brain barrier breakdown by NOX5 limits postreperfusion benefit in stroke. JO - J. Clin. Invest. VL - 129 IS - 4 PB - Amer Soc Clinical Investigation Inc PY - 2019 SN - 0021-9738 ER - TY - JOUR AB - The metabolic syndrome (MetS) encompasses medical conditions such as obesity, hyperglycemia, high blood pressure, and dyslipidemia that are major drivers for the ever-increasing prevalence of type 2 diabetes, cardiovascular diseases, and certain types of cancer. At the core of clinical strategies against the MetS is weight loss, induced by bariatric surgery, lifestyle changes based on calorie reduction and exercise, or pharmacology. This Review summarizes the past, current, and future efforts of targeting the MetS by pharmacological agents. Major emphasis is given to drugs that target the CNS as a key denominator for obesity and its comorbid sequelae. AU - Stemmer, K. AU - Müller, T.D. AU - DiMarchi, R.D.* AU - Pfluger, P.T. AU - Tschöp, M.H. C1 - 56709 C2 - 47252 CY - 2015 Manchester Rd, Ann Arbor, Mi 48104 Usa SP - 4058-4071 TI - CNS-targeting pharmacological interventions for the metabolic syndrome. JO - J. Clin. Invest. VL - 130 IS - 10 PB - Amer Soc Clinical Investigation Inc PY - 2019 SN - 0021-9738 ER - TY - JOUR AB - T cell therapy is a promising means to treat chronic hepatitis B virus (HBV) infection and HBV-associated hepatocellular carcinoma. T cells engineered to express an HBV-specific T cell receptor (TCR) may cure an HBV infection upon adoptive transfer. We investigated the therapeutic potential and safety of T cells stably expressing high-affinity HBV envelope- or core-specific TCRs recognizing European and Asian HLA-A2 subtypes. Both CD8(+) and CD4(+) T cells from healthy donors and patients with chronic hepatitis B became polyfunctional effector cells when grafted with HBV-specific TCRs and eliminated HBV from infected HepG2-NTCP cell cultures. A single transfer of TCR-grafted T cells into HBV-infected, humanized mice controlled HBV infection, and virological markers declined by 4 to 5 log or below the detection limit. Engineered T cells specifically cleared infected hepatocytes without damaging noninfected cells when, as in a typical clinical setting, only a minority of hepatocytes were infected. Cell death was compensated by hepatocyte proliferation, and alanine amino transferase levels peaking between days 5 and 7 normalized again thereafter. Cotreatment with the entry inhibitor myrcludex B ensured long-term control of HBV infection. Thus, T cells stably transduced with highly functional TCRs have the potential to mediate clearance of HBV-infected cells, causing limited liver injury. AU - Wisskirchen, K. AU - Kah, J.* AU - Malo, A. AU - Asen, T. AU - Volz, T.* AU - Allweiss, L.* AU - Wettengel, J.M.* AU - Lütgehetmann, M.* AU - Urban, S.* AU - Bauer, T. AU - Dandri, M.* AU - Protzer, U. C1 - 56017 C2 - 46695 CY - 2015 Manchester Rd, Ann Arbor, Mi 48104 Usa SP - 2932-2945 TI - T cell receptor grafting allows virological control of hepatitis B virus infection. JO - J. Clin. Invest. VL - 130 IS - 7 PB - Amer Soc Clinical Investigation Inc PY - 2019 SN - 0021-9738 ER - TY - JOUR AB - High-risk neuroblastoma is a devastating malignancy with very limited therapeutic options. Here, we identify withaferin A (WA) as a natural ferroptosis-inducing agent in neuroblastoma, which acts through a novel double-edged mechanism. WA dose-dependently either activates the nuclear factor-like 2 pathway through targeting of Kelch-like ECH-associated protein 1 (noncanonical ferroptosis induction) or inactivates glutathione peroxidase 4 (canonical ferroptosis induction). Noncanonical ferroptosis induction is characterized by an increase in intracellular labile Fe(II) upon excessive activation of heme oxygenase-1, which is sufficient to induce ferroptosis. This double-edged mechanism might explain the superior efficacy of WA as compared with etoposide or cisplatin in killing a heterogeneous panel of high-risk neuroblastoma cells, and in suppressing the growth and relapse rate of neuroblastoma xenografts. Nano-targeting of WA allows systemic application and suppressed tumor growth due to an enhanced accumulation at the tumor site. Collectively, our data propose a novel therapeutic strategy to efficiently kill cancer cells by ferroptosis. AU - Hassannia, B.* AU - Wiernicki, B.* AU - Ingold, I. AU - Qu, F.* AU - Van Herck, S.* AU - Tyurina, Y.Y.* AU - Bayir, H.* AU - Abhari, B.A.* AU - Angeli, J.P.F.* AU - Choi, S.M.* AU - Meul, E.* AU - Heyninck, K.* AU - Declerck, K.* AU - Chirumamilla, C.S.* AU - Lahtela-Kakkonen, M.* AU - Van Camp, G.* AU - Krysko, D.V.* AU - Ekert, P.G.* AU - Fulda, S.* AU - De Geest, B.G.* AU - Conrad, M. AU - Kagan, V.E.* AU - Berghe, W.V.* AU - Vandenabeele, P.* AU - Berghe, T.V.* C1 - 53697 C2 - 44956 CY - 2015 Manchester Rd, Ann Arbor, Mi 48104 Usa SP - 3341-3355 TI - Nano-targeted induction of dual ferroptotic mechanisms eradicates high-risk neuroblastoma. JO - J. Clin. Invest. VL - 128 IS - 8 PB - Amer Soc Clinical Investigation Inc PY - 2018 SN - 0021-9738 ER - TY - JOUR AB - BACKGROUND: Dietary intake of saturated fat is a likely contributor to nonalcoholic fatty liver disease (NAFLD) and insulin resistance, but the mechanisms that initiate these abnormalities in humans remain unclear. We examined the effects of a single oral saturated fat load on insulin sensitivity, hepatic glucose metabolism, and lipid metabolism in humans. Similarly, initiating mechanisms were examined after an equivalent challenge in mice. METHODS: Fourteen lean, healthy individuals randomly received either palm oil (PO) or vehicle (VCL). Hepatic metabolism was analyzed using in vivo 13C/31P/1H and ex vivo 2H magnetic resonance spectroscopy before and during hyperinsulinemic-euglycemic clamps with isotope dilution. Mice underwent identical clamp procedures and hepatic transcriptome analyses. RESULTS: PO administration decreased whole-body, hepatic, and adipose tissue insulin sensitivity by 25%, 15%, and 34%, respectively. Hepatic triglyceride and ATP content rose by 35% and 16%, respectively. Hepatic gluconeogenesis increased by 70%, and net glycogenolysis declined by 20%. Mouse transcriptomics revealed that PO differentially regulates predicted upstream regulators and pathways, including LPS, members of the TLR and PPAR families, NF-κB, and TNF-related weak inducer of apoptosis (TWEAK). CONCLUSION: Saturated fat ingestion rapidly increases hepatic lipid storage, energy metabolism, and insulin resistance. This is accompanied by regulation of hepatic gene expression and signaling that may contribute to development of NAFLD.REGISTRATION. ClinicalTrials.gov NCT01736202. FUNDING: Germany: Ministry of Innovation, Science, and Research North Rhine-Westfalia, German Federal Ministry of Health, Federal Ministry of Education and Research, German Center for Diabetes Research, German Research Foundation, and German Diabetes Association. Portugal: Portuguese Foundation for Science and Technology, FEDER - European Regional Development Fund, Portuguese Foundation for Science and Technology, and Rede Nacional de Ressonância Magnética Nuclear.   AU - Álvarez Hernández, E.* AU - Kahl, S.* AU - Seelig, A. AU - Begovatz, P.* AU - Irmler, M. AU - Kupriyanova, Y.* AU - Nowotny, J.* AU - Nowotny, P.* AU - Herder, C.* AU - Barosa, C.* AU - Carvalho, F.P.* AU - Rozman, J. AU - Neschen, S. AU - Jones, J.G.* AU - Beckers, J. AU - Hrabě de Angelis, M. AU - Roden, M.* C1 - 50426 C2 - 42386 CY - Ann Arbor SP - 695-708 TI - Acute dietary fat intake initiates alterations in energy metabolism and insulin resistance. JO - J. Clin. Invest. VL - 127 IS - 2 PB - Amer Soc Clinical Investigation Inc PY - 2017 SN - 0021-9738 ER - TY - JOUR AB - Dysregulated adipocyte physiology leads to imbalanced energy storage, obesity, and associated diseases, imposing a costly burden on current health care. Cannabinoid receptor type-1 (CB1) plays a crucial role in controlling energy metabolism through central and peripheral mechanisms. In this work, adipocyte-specific inducible deletion of the CB1 gene (Ati-CB1- KO) was sufficient to protect adult mice from diet-induced obesity and associated metabolic alterations and to reverse the phenotype in already obese mice. Compared with controls, Ati-CB1-KO mice showed decreased body weight, reduced total adiposity, improved insulin sensitivity, enhanced energy expenditure, and fat depot-specific cellular remodeling toward lowered energy storage capacity and browning of white adipocytes. These changes were associated with an increase in alternatively activated macrophages concomitant with enhanced sympathetic tone in adipose tissue. Remarkably, these alterations preceded the appearance of differences in body weight, highlighting the causal relation between the loss of CB1 and the triggering of metabolic reprogramming in adipose tissues. Finally, the lean phenotype of Ati-CB1-KO mice and the increase in alternatively activated macrophages in adipose tissue were also present at thermoneutral conditions. Our data provide compelling evidence for a crosstalk among adipocytes, immune cells, and the sympathetic nervous system (SNS), wherein CB1 plays a key regulatory role. AU - De Azua, I.R.* AU - Mancini, G.M.* AU - Srivastava, R.K.* AU - Rey, A.A.* AU - Cardinal, P.* AU - Tedesco, L.* AU - Zingaretti, C.M.* AU - Sassmann, A.* AU - Quarta, C. AU - Schwitter, C.* AU - Conrad, A.* AU - Wettschureck, N.* AU - Vemuri, V.K.* AU - Makriyannis, A.* AU - Hartwig, J.H.* AU - Mendez-Lago, M.* AU - Bindila, L.* AU - Monory, K.* AU - Giordano, A.* AU - Cinti, S.* AU - Marsicano, G.* AU - Offermanns, S.* AU - Nisoli, E.* AU - Pagotto, U.* AU - Cota, D.* AU - Lutz, B.* C1 - 52326 C2 - 43878 SP - 4148-4162 TI - Adipocyte cannabinoid receptor CB1 regulates energy homeostasis and alternatively activated macrophages. JO - J. Clin. Invest. VL - 127 IS - 11 PY - 2017 SN - 0021-9738 ER - TY - JOUR AB - BACKGROUND. Understanding the genetic architecture of cardiac structure and function may help to prevent and treat heart disease. This investigation sought to identify common genetic variations associated with inter-individual variability in cardiac structure and function. METHODS. A GWAS meta-analysis of echocardiographic traits was performed, including 46,533 individuals from 30 studies (EchoGen consortium). The analysis included 16 traits of left ventricular (LV) structure, and systolic and diastolic function. RESULTS. The discovery analysis included 21 cohorts for structural and systolic function traits (n = 32,212) and 17 cohorts for diastolic function traits (n = 21,852). Replication was performed in 5 cohorts (n = 14,321) and 6 cohorts (n = 16,308), respectively. Besides 5 previously reported loci, the combined meta-analysis identified 10 additional genome-wide significant SNPs: rs12541595 near MTSS1 and rs10774625 in ATXN2 for LV end-diastolic internal dimension; rs806322 near KCNRG, rs4765663 in CACNA1C, rs6702619 near PALMD, rs7127129 in TMEM16A, rs11207426 near FGGY, rs17608766 in GOSR2, and rs17696696 in CFDP1 for aortic root diameter; and rs12440869 in IQCH for Doppler transmitral A-wave peak velocity. Findings were in part validated in other cohorts and in GWAS of related disease traits. The genetic loci showed associations with putative signaling pathways, and with gene expression in whole blood, monocytes, and myocardial tissue. CONCLUSION. The additional genetic loci identified in this large meta-analysis of cardiac structure and function provide insights into the underlying genetic architecture of cardiac structure and warrant follow-up in future functional studies. AU - Wild, P.S.* AU - Felix, J.F.* AU - Schillert, A.* AU - Teumer, A.* AU - Chen, M.* AU - Leening, M.J.G.* AU - Voelker, U.* AU - Grossmann, V.* AU - Brody, J.A.* AU - Irvin, M.R.* AU - Shah, S.J.* AU - Pramana, S.* AU - Lieb, W.* AU - Schmidt, R.* AU - Stanton, A.V.* AU - Malzahn, D.* AU - Smith, A.V.* AU - Sundström, J.* AU - Minelli, C.* AU - Ruggiero, D.* AU - Lyytikäinen, L.-P.* AU - Tiller, D.* AU - Smith, J.G.* AU - Monnereau, C.* AU - Di Tullio, M.R.* AU - Musani, S.K.* AU - Morrison, A.C.* AU - Pers, T.H.* AU - Morley, M.* AU - Kleber, M.E.* AU - Aragam, J.* AU - Benjamin, E.J.* AU - Bis, J.C.* AU - Bisping, E.* AU - Broeckel, U.* AU - Cheng, S.* AU - Deckers, J.W.* AU - Del Greco, F.* AU - Edelmann, F.* AU - Fornage, M.* AU - Franke, L.* AU - Friedrich, N.* AU - Harris, T.B.* AU - Hofer, E.* AU - Hofman, A.* AU - Huang, J.* AU - Hughes, A.D.* AU - Kähönen, M.* AU - Kruppa, J.* AU - Lackner, K.J.* AU - Lannfelt, L.* AU - Laskowski, R.A.* AU - Launer, L.J.* AU - Leosdottir, M.* AU - Lin, H.* AU - Lindgren, C.M.* AU - Loley, C.* AU - MacRae, C.A.* AU - Mascalzoni, D.* AU - Mayet, J.* AU - Medenwald, D.* AU - Morris, A.P.* AU - Mueller, C.* AU - Müller-Nurasyid, M. AU - Nappo, S.* AU - Nilsson, P.M.* AU - Nuding, S.* AU - Nutile, T.* AU - Peters, A. AU - Pfeufer, A. AU - Pietzner, D.* AU - Pramstaller, P.P.* AU - Raitakari, O.T.* AU - Rice, K.M.* AU - Rivadeneira, F.* AU - Rotter, J.I.* AU - Ruohonen, S.T.* AU - Sacco, R.L.* AU - Samdarshi, T.E.* AU - Schmidt, H.* AU - Sharp, A.S.P.* AU - Shields, D.C.* AU - Sorice, R.* AU - Sotoodehnia, N.* AU - Stricker, B.H.* AU - Surendran, P.* AU - Thom, S.* AU - Toeglhofer, A.M.* AU - Uitterlinden, A.G.* AU - Wachter, R.* AU - Voelzke, H.* AU - Ziegler, A.* AU - Muenzel, T.* AU - Maerz, W.* AU - Cappola, T.P.* AU - Hirschhorn, J.N.* AU - Mitchell, G.F.* AU - Smith, N.L.* AU - Fox, E.R.* AU - Dueker, N.D.* AU - Jaddoe, V.W.V.* AU - Melander, O.* AU - Russ, M.* AU - Lehtimäki, T.* AU - Ciullo, M.* AU - Hicks, A.A.* AU - Lind, L.* AU - Gudnason, V.* AU - Pieske, B.* AU - Barron, A.J.* AU - Zweiker, R.* AU - Schunkert, H.* AU - Ingelsson, E.* AU - Liu, K.* AU - Arnett, D.K.* AU - Psaty, B.M.* AU - Blankenberg, S.* AU - Larson, M.G.* AU - Felix, S.B.* AU - Franco, O.H.* AU - Zeller, T.* AU - Vasan, R.S.* AU - Doerr, M.* C1 - 51127 C2 - 42703 CY - Ann Arbor SP - 1798-1812 TI - Large-scale genome-wide analysis identifies genetic variants associated with cardiac structure and function. JO - J. Clin. Invest. VL - 127 IS - 5 PB - Amer Soc Clinical Investigation Inc PY - 2017 SN - 0021-9738 ER - TY - JOUR AB - Autonomous thyroid adenomas (ATAs) are a frequent cause of hyperthyroidism. Mutations in the genes encoding the TSH receptor (TSHR) or the Gs protein α subunit (GNAS) are found in approximately 70% of ATAs. The involvement of other genes and the pathogenesis of the remaining cases are presently unknown. Here, we performed whole-exome sequencing in 19 ATAs that were paired with normal DNA samples and identified a recurrent hot-spot mutation (c.1712A>G; p.Gln571Arg) in the enhancer of zeste homolog 1 (EZH1) gene, which codes for a catalytic subunit of the polycomb complex. Targeted screening in an independent cohort confirmed that this mutation occurs with high frequency (27%) in ATAs. EZH1 mutations were strongly associated with known (TSHR, GNAS) or presumed (adenylate cyclase 9 [ADCY9]) alterations in cAMP pathway genes. Furthermore, functional studies revealed that the p.Gln571Arg EZH1 mutation caused increased histone H3 trimethylation and increased proliferation of thyroid cells. In summary, this study revealed that a hot-spot mutation in EZH1 is the second most frequent genetic alteration in ATAs. The association between EZH1 and TSHR mutations suggests a 2-hit model for the pathogenesis of these tumors, whereby constitutive activation of the cAMP pathway and EZH1 mutations cooperate to induce the hyperproliferation of thyroid cells. AU - Calebiro, D.* AU - Grassi, E.S.* AU - Eszlinger, M.* AU - Ronchi, C.L.* AU - Godbole, A.* AU - Bathon, K.* AU - Guizzardi, F.* AU - de Filippis, T.* AU - Krohn, K.* AU - Jaeschke, H.* AU - Schwarzmayr, T. AU - Bircan, R.* AU - Gozu, H.I.* AU - Sancak, S.* AU - Niedziela, M.* AU - Strom, T.M. AU - Fassnacht, M.* AU - Persani, L.* AU - Paschke, R.* C1 - 49372 C2 - 41790 CY - Ann Arbor SP - 3383-3388 TI - Recurrent EZH1 mutations are a second hit in autonomous thyroid adenomas. JO - J. Clin. Invest. VL - 126 IS - 9 PB - Amer Soc Clinical Investigation Inc PY - 2016 SN - 0021-9738 ER - TY - JOUR AB - Interactions of diet, gut microbiota, and host genetics play important roles in the development of obesity and insulin resistance. Here, we have investigated the molecular links between gut microbiota, insulin resistance, and glucose metabolism in 3 inbred mouse strains with differing susceptibilities to metabolic syndrome using diet and antibiotic treatment. Antibiotic treatment altered intestinal microbiota, decreased tissue inflammation, improved insulin signaling in basal and stimulated states, and improved glucose metabolism in obesity- and diabetes-prone C57BL/6J mice on a high-fat diet (HFD). Many of these changes were reproduced by the transfer of gut microbiota from antibiotic-treated donors to germ-free or germ-depleted mice. These physiological changes closely correlated with changes in serum bile acids and levels of the antiinflammatory bile acid receptor Takeda G protein–coupled receptor 5 (TGR5) and were partially recapitulated by treatment with a TGR5 agonist. In contrast, antibiotic treatment of HFD-fed, obesity-resistant 129S1 and obesity-prone 129S6 mice did not improve metabolism, despite changes in microbiota and bile acids. These mice also failed to show a reduction in inflammatory gene expression in response to the TGR5 agonist. Thus, changes in bile acid and inflammatory signaling, insulin resistance, and glucose metabolism driven by an HFD can be modified by antibiotic-induced changes in gut microbiota; however, these effects depend on important interactions with the host’s genetic background and inflammatory potential. AU - Fujisaka, S.* AU - Ussar, S. AU - Clish, C.B.* AU - Devkota, S.* AU - Dreyfuss, J.M.* AU - Sakaguchi, M.* AU - Soto, M.* AU - Konsishi, M.* AU - Softic, S.* AU - Altindis, E.* AU - Li, N.* AU - Gerber, G.* AU - Bry, L.* AU - Kahn, R.* C1 - 50080 C2 - 42004 CY - Ann Arbor SP - 4430-4443 TI - Antibiotic effects on gut microbiota and metabolism are host dependent. JO - J. Clin. Invest. VL - 126 IS - 12 PB - Amer Soc Clinical Investigation Inc PY - 2016 SN - 0021-9738 ER - TY - JOUR AB - Tumor suppression that is mediated by oncogene-induced senescence (OIS) is considered to function as a safeguard during development of pancreatic ductal adenocarcinoma (PDAC). However, the mechanisms that regulate OIS in PDAC are poorly understood. Here, we have determined that nuclear RelA reinforces OIS to inhibit carcinogenesis in the Kras mouse model of PDAC. Inactivation of RelA accelerated pancreatic lesion formation in Kras mice by abrogating the senescence-associated secretory phenotype (SASP) gene transcription signature. Using genetic and pharmacological tools, we determined that RelA activation promotes OIS via elevation of the SASP factor CXCL1 (also known as KC), which activates CXCR2, during pancreatic carcinogenesis. In Kras mice, pancreas-specific inactivation of CXCR2 prevented OIS and was correlated with increased tumor proliferation and decreased survival. Moreover, reductions in CXCR2 levels were associated with advanced neoplastic lesions in tissue from human pancreatic specimens. Genetically disabling OIS in Kras mice caused RelA to promote tumor proliferation, suggesting a dual role for RelA signaling in pancreatic carcinogenesis. Taken together, our data suggest a pivotal role for RelA in regulating OIS in preneoplastic lesions and implicate the RelA/CXCL1/CXCR2 axis as an essential mechanism of tumor surveillance in PDAC. AU - Lesina, M.* AU - Wörmann, S.M.* AU - Morton, J.* AU - Diakopoulos, K.N.* AU - Korneeva, O.* AU - Wimmer, M.* AU - Einwächter, H.* AU - Sperveslage, J.* AU - Demir, I.E.* AU - Kehl, T.* AU - Saur, D.* AU - Sipos, B.* AU - Heikenwälder, M. AU - Steiner, J.M.* AU - Wang, T.C.* AU - Sansom, O.J.* AU - Schmid, R.M.* AU - Algül, H.* C1 - 49376 C2 - 41786 CY - Ann Arbor SP - 2919-2932 TI - RelA regulates CXCL1/CXCR2-dependent oncogene-induced senescence in murine Kras-driven pancreatic carcinogenesis. JO - J. Clin. Invest. VL - 126 IS - 8 PB - Amer Soc Clinical Investigation Inc PY - 2016 SN - 0021-9738 ER - TY - JOUR AB - In Wilson disease (WD), functional loss of ATPase copper-transporting β (ATP7B) impairs biliary copper excretion, leading to excessive copper accumulation in the liver and fulminant hepatitis. Current US Food and Drug Administration- and European Medicines Agency-approved pharmacological treatments usually fail to restore copper homeostasis in patients with WD who have progressed to acute liver failure, leaving liver transplantation as the only viable treatment option. Here, we investigated the therapeutic utility of methanobactin (MB), a peptide produced by Methylosinus trichosporium OB3b, which has an exceptionally high affinity for copper. We demonstrated that ATP7B-deficient rats recapitulate WD-associated phenotypes, including hepatic copper accumulation, liver damage, and mitochondrial impairment. Short-term treatment of these rats with MB efficiently reversed mitochondrial impairment and liver damage in the acute stages of liver copper accumulation compared with that seen in untreated ATP7B-deficient rats. This beneficial effect was associated with depletion of copper from hepatocyte mitochondria. Moreover, MB treatment prevented hepatocyte death, subsequent liver failure, and death in the rodent model. These results suggest that MB has potential as a therapeutic agent for the treatment of acute WD. AU - Lichtmannegger, J. AU - Leitzinger, C. AU - Wimmer, R.* AU - Schmitt, S. AU - Schulz, S. AU - Kabiri, Y. AU - Eberhagen, C. AU - Rieder, T.* AU - Janik, D. AU - Neff, F. AU - Straub, B.K.* AU - Schirmacher, P.* AU - DiSpirito, A.A.* AU - Bandow, N.* AU - Baral, B.S.* AU - Flatley, A. AU - Kremmer, E. AU - Denk, G.U.* AU - Reiter, F.P.* AU - Hohenester, S.* AU - Eckardt-Schupp, F. AU - Dencher, N.A.* AU - Adamski, J. AU - Sauer, V.* AU - Niemietz, C.* AU - Schmidt, H.H.* AU - Merle, U.* AU - Gotthardt, D.N.* AU - Kroemer, G.* AU - Weiss, K.H.* AU - Zischka, H. C1 - 48921 C2 - 41495 CY - Ann Arbor SP - 2721-2735 TI - Methanobactin reverses acute liver failure in a rat model of Wilson disease. JO - J. Clin. Invest. VL - 126 IS - 7 PB - Amer Soc Clinical Investigation Inc PY - 2016 SN - 0021-9738 ER - TY - JOUR AB - Dietary protein intake is linked to an increased incidence of type 2 diabetes (T2D). Although dietary protein dilution (DPD) can slow the progression of some aging-related disorders, whether this strategy affects the development and risk for obesity-associated metabolic disease such as T2D is unclear. Here, we determined that DPD in mice and humans increases serum markers of metabolic health. In lean mice, DPD promoted metabolic inefficiency by increasing carbohydrate and fat oxidation. In nutritional and polygenic murine models of obesity, DPD prevented and curtailed the development of impaired glucose homeostasis independently of obesity and food intake. DPD-mediated metabolic inefficiency and improvement of glucose homeostasis were independent of uncoupling protein 1 (UCP1), but required expression of liver-derived fibroblast growth factor 21 (FGF21) in both lean and obese mice. FGF21 expression and secretion as well as the associated metabolic remodeling induced by DPD also required induction of liver-integrated stress response-driven nuclear protein 1 (NUPR1). Insufficiency of select nonessential amino acids (NEAAs) was necessary and adequate for NUPR1 and subsequent FGF21 induction and secretion in hepatocytes in vitro and in vivo. Taken together, these data indicate that DPD promotes improved glucose homeostasis through an NEAA insufficiency-induced liver NUPR1/FGF21 axis. AU - Maida, A. AU - Zota, A. AU - Sjøberg, K.A.* AU - Schumacher, J.* AU - Sijmonsma, T.P.* AU - Pfenninger, A.* AU - Christensen, M.M.* AU - Gantert, T.* AU - Fuhrmeister, J.* AU - Rothermel, U.* AU - Schmoll, D.* AU - Heikenwälder, M.* AU - Iovanna, J.L.* AU - Stemmer, K. AU - Kiens, B.* AU - Herzig, S. AU - Rose, A.J.* C1 - 49369 C2 - 41793 CY - Ann Arbor SP - 3263-3278 TI - A liver stress-endocrine nexus promotes metabolic integrity during dietary protein dilution. JO - J. Clin. Invest. VL - 126 IS - 9 PB - Amer Soc Clinical Investigation Inc PY - 2016 SN - 0021-9738 ER - TY - JOUR AB - The genetic disorder Kabuki syndrome (KS) is characterized by developmental delay and congenital anomalies. Dominant mutations in the chromatin regulators lysine (K)-specific methyltransferase 2D (KMT2D) (also known as MLL2) and lysine (K)-specific demethylase 6A (KDM6A) underlie the majority of cases. Although the functions of these chromatin-modifying proteins have been studied extensively, the physiological systems regulated by them are largely unknown. Using whole-exome sequencing, we identified a mutation in RAP1A that was converted to homozygosity as the result of uniparental isodisomy (UPD) in a patient with KS and a de novo, dominant mutation in RAP1B in a second individual with a KS-like phenotype. We elucidated a genetic and functional interaction between the respective KS-associated genes and their products in zebrafish models and patient cell lines. Specifically, we determined that dysfunction of known KS genes and the genes identified in this study results in aberrant MEK/ERK signaling as well as disruption of F-actin polymerization and cell intercalation. Moreover, these phenotypes could be rescued in zebrafish models by rebalancing MEK/ERK signaling via administration of small molecule inhibitors of MEK. Taken together, our studies suggest that the KS pathophysiology overlaps with the RASopathies and provide a potential direction for treatment design. AU - Bögershausen, N.* AU - Tsai, I.C.* AU - Pohl, E.* AU - Kiper, P.* AU - Beleggia, F.* AU - Percin, E.F.* AU - Keupp, K.* AU - Matchan, A.* AU - Milz, E.* AU - Alanay, Y.* AU - Kayserili, H.* AU - Liu, A.H.* AU - Banka, S.* AU - Kranz, A.* AU - Zenker, M.* AU - Wieczorek, D.* AU - Elcioğlu, N.H.* AU - Prontera, P.* AU - Lyonnet, S.* AU - Meitinger, T. AU - Stewart, A.F.* AU - Donnai, D.* AU - Strom, T.M. AU - Boduroglu, K.* AU - Yigit, G.* AU - Li, Y.* AU - Katsanis, N.* AU - Wollnik, B.* C1 - 46768 C2 - 37805 SP - 3585-3599 TI - RAP1-mediated MEK/ERK pathway defects in Kabuki syndrome. JO - J. Clin. Invest. VL - 125 IS - 9 PY - 2015 SN - 0021-9738 ER - TY - JOUR AB - Mast cells (MCs) have been identified in various tumors; however, the role of these cells in tumorigenesis remains controversial. Here, we quantified MCs in human and murine malignant pleural effusions (MPEs) and evaluated the fate and function of these cells in MPE development. Evaluation of murine MPE-competent lung and colon adenocarcinomas revealed that these tumors actively attract and subsequently degranulate MCs in the pleural space by elaborating CCL2 and osteopontin. MCs were required for effusion development, as MPEs did not form in mice lacking MCs, and pleural infusion of MCs with MPE-incompetent cells promoted MPE formation. Once homed to the pleural space, MCs released tryptase AB1 and IL-1 beta, which in turn induced pleural vasculature leakiness and triggered NF-kappa B activation in pleural tumor cells, thereby fostering pleural fluid accumulation and tumor growth. Evaluation of human effusions revealed that MCs are elevated in MPEs compared with benign effusions. Moreover, MC abundance correlated with MPE formation in a human cancer cell-induced effusion model. Treatment of mice with the c-KIT inhibitor imatinib mesylate limited effusion precipitation by mouse and human adenbcarcinoma cells. Together, the results of this study indicate that MCs are required for MPE formation and suggest that MC-dependent effusion formation is therapeutically addressable. AU - Giannou, A.D.* AU - Marazioti, A.* AU - Spella, M.* AU - Kanellakis, N.I.* AU - Apostolopoulou, H.* AU - Psallidas, L.* AU - Prijovich, Z.M.* AU - Vreka, M.* AU - Zazara, D.E.* AU - Lilis, L.* AU - Papaleonidopoulos, V.* AU - Kairi, C.A.* AU - Patmanidi, A.L.* AU - Giopanou, J.* AU - Spiropoulou, N.* AU - Harokopos, V.* AU - Aidinis, V.* AU - Spyratos, D.* AU - Teliousi, S.* AU - Papadaki, H.* AU - Taraviras, S.* AU - Snyder, L.A.* AU - Eickelberg, O. AU - Kardamakis, D.* AU - Iwakura, V.* AU - Feyerabend, T.B.* AU - Rodewald, H.R.* AU - Kalomenidis, L.* AU - Blackwell, T.S.* AU - Agalloti, T.* AU - Stathopoulos, G.T. C1 - 45421 C2 - 37377 CY - Ann Arbor SP - 2317-2334 TI - Mast cells mediate malignant pleural effusion formation. JO - J. Clin. Invest. VL - 125 IS - 6 PB - Amer Soc Clinical Investigation Inc PY - 2015 SN - 0021-9738 ER - TY - JOUR AB - Ductular reactions (DRs) are observed in virtually all forms of human liver disease; however, the histogenesis and function of DRs in liver injury are not entirely understood. It is widely believed that DRs contain bipotential liver progenitor cells (LPCs) that serve as an emergency cell pool to regenerate both cholangiocytes and hepatocytes and may eventually give rise to hepatocellular carcinoma (HCC). Here, we used a murine model that allows highly efficient and specific lineage labeling of the biliary compartment to analyze the histogenesis of DRs and their potential contribution to liver regeneration and carcinogenesis. In multiple experimental and genetic liver injury models, biliary cells were the predominant precursors of DRs but lacked substantial capacity to produce new hepatocytes, even when liver injuries were prolonged up to 12 months. Genetic modulation of NOTCH and/or WNT/β-catenin signaling within lineage-tagged DRs impaired DR expansion but failed to redirect DRs from biliary differentiation toward the hepatocyte lineage. Further, lineage-labeled DRs did not produce tumors in genetic and chemical HCC mouse models. In summary, we found no evidence in our system to support mouse biliary-derived DRs as an LPC pool to replenish hepatocytes in a quantitatively relevant way in injury or evidence that DRs give rise to HCCs. AU - Jors, S.* AU - Jeliazkova, P.* AU - Ringelhan, M. AU - Thalhammer, J.* AU - Dürl, S.* AU - Ferrer, J.* AU - Sander, M.* AU - Heikenwälder, M. AU - Schmid, R.M.* AU - Siveke, J.T.* AU - Geisler, F.* C1 - 44743 C2 - 36990 CY - Ann Arbor SP - 2445-2457 TI - Lineage fate of ductular reactions in liver injury and carcinogenesis. JO - J. Clin. Invest. VL - 125 IS - 6 PB - Amer Soc Clinical Investigation Inc PY - 2015 SN - 0021-9738 ER - TY - JOUR AB - In many organs, including the intestine and skin, cancers originate from cells of the stem or progenitor compartment. Despite its nomenclature, the cellular origin of hepatocellular carcinoma (HCC) remains elusive. In contrast to most organs, the liver lacks a defined stem cell population for organ maintenance. Previous studies suggest that both hepatocytes and facultative progenitor cells within the biliary compartment are capable of generating HCC. As HCCs with a progenitor signature carry a worse prognosis, understanding the origin of HCC is of clinical relevance. Here, we used complementary fate-tracing approaches to label the progenitor/biliary compartment and hepatocytes in murine hepatocarcinogenesis. In genotoxic and genetic models, HCCs arose exclusively from hepatocytes but never from the progenitor/biliary compartment. Cytokeratin 19-, A6- and α-fetoprotein-positive cells within tumors were hepatocyte derived. In summary, hepatocytes represent the cell of origin for HCC in mice, and a progenitor signature does not reflect progenitor origin, but dedifferentiation of hepatocyte-derived tumor cells. AU - Mu, X.* AU - Español-Suñer, R.* AU - Mederacke, I.* AU - Affò, S.* AU - Manco, R.* AU - Sempoux, C.* AU - Lemaigre, F.P.* AU - Adili, A. AU - Yuan, D.T. AU - Weber, A.* AU - Unger, K. AU - Heikenwälder, M. AU - Leclercq, I.A.* AU - Schwabe, R.F.* C1 - 46935 C2 - 39067 SP - 3891-3903 TI - Hepatocellular carcinoma originates from hepatocytes and not from the progenitor/biliary compartment. JO - J. Clin. Invest. VL - 125 IS - 10 PY - 2015 SN - 0021-9738 ER - TY - JOUR AB - Type 2 diabetes mellitus (T2DM) is a worldwide heath problem that is characterized by insulin resistance and the eventual loss of β cell function. As recent studies have shown that loss of ribosomal protein (RP) S6 kinase 1 (S6K1) increases systemic insulin sensitivity, S6K1 inhibitors are being pursued as potential agents for improving insulin resistance. Here we found that S6K1 deficiency in mice also leads to decreased β cell growth, intrauterine growth restriction (IUGR), and impaired placental development. IUGR is a common complication of human pregnancy that limits the supply of oxygen and nutrients to the developing fetus, leading to diminished embryonic β cell growth and the onset of T2DM later in life. However, restoration of placental development and the rescue of IUGR by tetraploid embryo complementation did not restore β cell size or insulin levels in S6K1-/- embryos, suggesting that loss of S6K1 leads to an intrinsic β cell lesion. Consistent with this hypothesis, reexpression of S6K1 in β cells of S6K1-/- mice restored embryonic β cell size, insulin levels, glucose tolerance, and RPS6 phosphorylation, without rescuing IUGR. Together, these data suggest that a nutrient-mediated reduction in intrinsic β cell S6K1 signaling, rather than IUGR, during fetal development may underlie reduced β cell growth and eventual development of T2DM later in life. AU - Um, S.H.* AU - Sticker-Jantscheff, M.* AU - Chau, G.C.* AU - Vintersten, K.* AU - Mueller, M.* AU - Gangloff, Y.G.* AU - Adams, R.H.* AU - Spetz, J.F.* AU - Elghazi, L.* AU - Pfluger, P.T. AU - Pende, M.* AU - Bernal-Mizrachi, E.* AU - Tauler, A.* AU - Tschöp, M.H. AU - Thomas, G.* AU - Kozma, S.C.* C1 - 45601 C2 - 37389 CY - Ann Arbor SP - 2736-2747 TI - S6K1 controls pancreatic β cell size independently of intrauterine growth restriction. JO - J. Clin. Invest. VL - 125 IS - 7 PB - Amer Soc Clinical Investigation Inc PY - 2015 SN - 0021-9738 ER - TY - JOUR AB - Caspase-3-mediated spontaneous death in neutrophils is a prototype of programmed cell death and is critical for modulating physiopathological inflammatory responses; however, the underlying regulatory pathways remain ill defined. Here we determined that in aging neutrophils, the cleavage and activation of caspase-3 is independent of the canonical caspase-8- or caspase-9-mediated pathway. Instead, caspase-3 activation was mediated by serine protease proteinase 3 (PR3), which is present in the cytosol of aging neutrophils. Specifically, PR3 cleaved procaspase-3 at a site upstream of the canonical caspase-9 cleavage site. In mature neutrophils, PR3 was sequestered in granules and released during aging via lysosomal membrane permeabilization (LMP), leading to procaspase-3 cleavage and apoptosis. Pharmacological inhibition or knockdown of PR3 delayed neutrophil death in vitro and consistently delayed neutrophil death and augmented neutrophil accumulation at sites of inflammation in a murine model of peritonitis. Adoptive transfer of both WT and PR3-deficient neutrophils revealed that the delayed death of neutrophils lacking PR3 is due to an altered intrinsic apoptosis/survival pathway, rather than the inflammatory microenvironment. The presence of the suicide protease inhibitor SERPINB1 counterbalanced the protease activity of PR3 in aging neutrophils, and deletion of Serpinb1 accelerated neutrophil death. Taken together, our results reveal that PR3-mediated caspase-3 activation controls neutrophil spontaneous death. AU - Loison, F.* AU - Zhu, H.* AU - Karatepe, K.* AU - Kasorn, A.* AU - Liu, P.* AU - Ye, K.* AU - Zhou, J.* AU - Cao, S.* AU - Gong, H.* AU - Jenne, D. AU - Remold-O'Donnell, E.* AU - Xu, Y.* AU - Luo, H.R.* C1 - 32511 C2 - 35084 CY - Ann Arbor SP - 4445-4458 TI - Proteinase 3-dependent caspase-3 cleavage modulates neutrophil death and inflammation. JO - J. Clin. Invest. VL - 124 IS - 10 PB - Amer Soc Clinical Investigation Inc PY - 2014 SN - 0021-9738 ER - TY - JOUR AB - Who among us hasn't fantasized about a diet that allows ingestion of a surfeit of calories that are burned off effortlessly by ramping up energy expenditure? In this issue of the JCI, research led by Christopher Morrison suggests that this dream may become a reality; however, a complete understanding of the molecular interface that connects nutrient choices with our cellular metabolism will be required. Laeger et al. show that the expression and secretion of the weight-reducing hormone fibroblast growth factor 21 (FGF21) is regulated by dietary proteins and not, as has been heretofore assumed, simply triggered by reduced caloric intake. This study not only sheds new light on the role of FGF21 in systems metabolism, but also on the ways our bodies cope with the ever-changing availability of different dietary macronutrients. AU - Müller, T.D. AU - Tschöp, M.H. C1 - 31970 C2 - 34919 CY - Ann Arbor SP - 3691-3693 TI - Play down protein to play up metabolism? JO - J. Clin. Invest. VL - 124 IS - 9 PB - Amer Soc Clinical Investigation Inc PY - 2014 SN - 0021-9738 ER - TY - JOUR AB - Identification of single-gene causes of steroid-resistant nephrotic syndrome (SRNS) has furthered the understanding of the pathogenesis of this disease. Here, using a combination of homozygosity mapping and whole human exome resequencing, we identified mutations in the aarF domain containing kinase 4 (ADCK4) gene in 15 individuals with SRNS from 8 unrelated families. ADCK4 was highly similar to ADCK3, which has been shown to participate in coenzyme Q(10) (CoQ(10)) biosynthesis. Mutations in ADCK4 resulted in reduced COQ(10). levels and reduced mitochondrial respiratory enzyme activity in cells isolated from individuals with SRNS and transformed lymphoblasts. Knockdown of adck4 in zebrafish and Drosophila recapitulated nephrotic syndrome-associated phenotypes. Furthermore, ADCK4 was expressed in glomerular podocytes and partially localized to podocyte mitochondria and foot processes in rat kidneys and cultured human podocytes. In human podocytes, ADCK4 interacted with members of the CoQ(10) biosynthesis pathway, including COQ6, which has been linked with SRNS and COQ7. Knockdown of ADCK4 in podocytes resulted in decreased migration, which was reversed by CoQ(10) addition. Interestingly, a patient with SRNS with a homozygous ADCK4 frameshift mutation had partial remission following CoQ(10) treatment. These data indicate that individuals with SRNS with mutations in ADCK4 or other genes that participate in CoQ(10) biosynthesis may be treatable with CoQ(10). AU - Ashraf, S.* AU - Gee, H.Y.* AU - Woerner, S.* AU - Xie, L.T.X.* AU - Vega-Warner, V.* AU - Lovric, S.* AU - Fang, H.* AU - Song, X.W.* AU - Cattran, D.C.* AU - Avila-Casado, C.* AU - Paterson, A.D.* AU - Nitschke, P.* AU - Bole-Feysot, C.* AU - Cochat, P.* AU - Esteve-Rudd, J.* AU - Haberberger, B. AU - Allen, S.J.* AU - Zhou, W.B.* AU - Airik, R.* AU - Otto, E.A.* AU - Barua, M.* AU - Al-Hamed, M.H.* AU - Kari, J.A.* AU - Evans, J.* AU - Bierzynska, A.* AU - Saleem, M.A.* AU - Bockenhauer, D.* AU - Kleta, R.* AU - El Desoky, S.* AU - Hacihamdioglu, D.O.* AU - Gok, F.* AU - Washburn, J.* AU - Wiggins, R.C.* AU - Choi, M.* AU - Lifton, R.P.* AU - Levy, S.* AU - Han, Z.G.* AU - Salviati, L.* AU - Prokisch, H. AU - Williams, D.S.* AU - Pollak, M.* AU - Clarke, C.F.* AU - Pei, Y.* AU - Antignac, C.* AU - Hildebrandt, F.* C1 - 28840 C2 - 34590 SP - 5179-5189 TI - ADCK4 mutations promote steroid-resistant nephrotic syndrome through CoQ10 biosynthesis disruption. JO - J. Clin. Invest. VL - 123 IS - 12 PB - Amer. Soc. Clinical Investigation PY - 2013 SN - 0021-9738 ER - TY - JOUR AB - The suppression of tumorigenicity 2/IL-33 (ST2/IL-33) pathway has been implicated in several immune and inflammatory diseases. ST2 is produced as 2 isoforms. The membrane-bound isoform (ST2L) induces an immune response when bound to its ligand, IL-33. The other isoform is a soluble protein (sST2) that is thought to be a decoy receptor for IL-33 signaling. Elevated sST2 levels in serum are associated with an increased risk for cardiovascular disease. We investigated the determinants of sST2 plasma concentrations in 2,991 Framingham Offspring Cohort participants. While clinical and environmental factors explained some variation in sST2 levels, much of the variation in sST2 production was driven by genetic factors. In a genome-wide association study (GWAS), multiple SNPs within IL1RL1 (the gene encoding ST2) demonstrated associations with sST2 concentrations. Five missense variants of IL1RL1 correlated with higher sST2 levels in the GWAS and mapped to the intracellular domain of ST2, which is absent in sST2. In a cell culture model, IL1RL1 missense variants increased sST2 expression by inducing IL-33 expression and enhancing IL-33 responsiveness (via ST2L). Our data suggest that genetic variation in IL1RL1 can result in increased levels of sST2 and alter immune and inflammatory signaling through the ST2/IL-33 pathway. AU - Ho, J.E.* AU - Chen, W.Y.* AU - Chen, M.H.* AU - Larson, M.G.* AU - McCabe, E.L.* AU - Cheng, S.* AU - Ghorbani, A.* AU - Coglianese, E.* AU - Emilsson, V.* AU - Johnson, A.D.* AU - Walter, S.* AU - Franceschini, N.* AU - O'Donnell, C.J.* AU - CARDIoGRAM Consortium (Wichmann, H.-E. AU - Döring, A. AU - Illig, T. AU - Meisinger, C. AU - Peters, A. AU - Meitinger, T. AU - Klopp, N.) AU - CHARGE Inflammation Working Group (*) AU - Dehghan, A.* AU - Lu, C.* AU - Levy, D.* AU - Newton-Cheh, C.* AU - CHARGE Consortium Heart Failure Group (*) AU - Lin, H.* AU - Felix, J.F.* AU - Schreiter, E.R.* AU - Vasan, R.S.* AU - Januzzi, J.L. Jr.* AU - Lee, R.T.* AU - Wang, T.J.* AU - Assimes, T.L.* AU - Deloukas, P.* AU - Erdmann, J.* AU - Holm, H.* AU - Kathiresan, S.* AU - König, I.R.* AU - McPherson, R.* AU - Reilly, M.P.* AU - Roberts, R.* AU - Samani, N.J.* AU - Schunkert, H.* AU - Stewart, A.F.* C1 - 28869 C2 - 33555 SP - 4208-4218 TI - Common genetic variation at the IL1RL1 locus regulates IL-33/ST2 signaling. JO - J. Clin. Invest. VL - 123 IS - 10 PY - 2013 SN - 0021-9738 ER - TY - JOUR AB - The scaffold protein p62 (sequestosome 1; SQSTM1) is an emerging key molecular link among the metabolic, immune, and proliferative processes of the cell. Here, we report that adipocyte-specific, but not CNS-, liver-, muscle-, or myeloid-specific p62-deficient mice are obese and exhibit a decreased metabolic rate caused by impaired nonshivering thermogenesis. Our results show that p62 regulates energy metabolism via control of mitochondrial function in brown adipose tissue (BAT). Accordingly, adipocyte-specific p62 deficiency led to impaired mitochondrial function, causing BAT to become unresponsive to β-adrenergic stimuli. Ablation of p62 leads to decreased activation of p38 targets, affecting signaling molecules that control mitochondrial function, such as ATF2, CREB, PGC1α, DIO2, NRF1, CYTC, COX2, ATP5β, and UCP1. p62 ablation in HIB1B and BAT primary cells demonstrated that p62 controls thermogenesis in a cell-autonomous manner, independently of brown adipocyte development or differentiation. Together, our data identify p62 as a novel regulator of mitochondrial function and brown fat thermogenesis. AU - Müller, T.D. AU - Lee, S.J.* AU - Jastroch, M. AU - Kabra, D. AU - Stemmer, K. AU - Aichler, M. AU - Abplanalp, B.* AU - Ananthakrishnan, G.* AU - Bhardwaj, N.* AU - Collins, S.* AU - Divanovic, S.* AU - Endele, M. AU - Finan, B. AU - Gao, Y.* AU - Habegger, K.M.* AU - Hembree, J.* AU - Heppner, K.M.* AU - Hofmann, S. AU - Holland, J.* AU - Küchler, D. AU - Kutschke, M. AU - Krishna, R.* AU - Lehti, M.* AU - Oelkrug, R.* AU - Ottaway, N.* AU - Perez-Tilve, D.* AU - Raver, C.* AU - Walch, A.K. AU - Schriever, S.C. AU - Speakman, J.* AU - Tseng, Y.H.* AU - Diaz-Meco, M.* AU - Pfluger, P.T. AU - Moscat, J.* AU - Tschöp, M.H. C1 - 11669 C2 - 30741 SP - 469-478 TI - P62 links β-adrenergic input to mitochondrial function and thermogenesis. JO - J. Clin. Invest. VL - 123 IS - 1 PB - American Society of Clinical Investigation Inc. PY - 2013 SN - 0021-9738 ER - TY - JOUR AB - Aging is a major risk factor for a large number of disorders and functional impairments. Therapeutic targeting of the aging process may therefore represent an innovative strategy in the quest for novel and broadly effective treatments against age-related diseases. The recent report of lifespan extension in mice treated with the FDA-approved mTOR inhibitor rapamycin represented the first demonstration of pharmacological extension of maximal lifespan in mammals. Longevity effects of rapamycin may, however, be due to rapamycin's effects on specific life-limiting pathologies, such as cancers, and it remains unclear if this compound actually slows the rate of aging in mammals. Here, we present results from a comprehensive, large-scale assessment of a wide range of structural and functional aging phenotypes, which we performed to determine whether rapamycin slows the rate of aging in male C57BL/6J mice. While rapamycin did extend lifespan, it ameliorated few studied aging phenotypes. A subset of aging traits appeared to be rescued by rapamycin. Rapamycin, however, had similar effects on many of these traits in young animals, indicating that these effects were not due to a modulation of aging, but rather related to aging-independent drug effects. Therefore, our data largely dissociate rapamycin's longevity effects from effects on aging itself. AU - Neff, F. AU - Flores-Dominguez, D.* AU - Ryan, D.P.* AU - Horsch, M. AU - Schröder, S.* AU - Adler, T. AU - Afonso, L.C. AU - Aguilar-Pimentel, J.A. AU - Becker, L. AU - Garrett, L. AU - Hans, W. AU - Hettich, M.M.* AU - Holtmeier, R.* AU - Hölter, S.M. AU - Moreth, K. AU - Prehn, C. AU - Puk, O. AU - Rácz, I.* AU - Rathkolb, B. AU - Rozman, J. AU - Naton, B. AU - Ordemann, R.* AU - Adamski, J. AU - Beckers, J. AU - Bekeredjian, R.* AU - Busch, D.H.* AU - Ehninger, G.* AU - Graw, J. AU - Höfler, H. AU - Klingenspor, M.* AU - Klopstock, T.* AU - Ollert, M.* AU - Stypmann, J.* AU - Wolf, E.* AU - Wurst, W. AU - Zimmer, A.* AU - Fuchs, H. AU - Gailus-Durner, V. AU - Hrabě de Angelis, M. AU - Ehniger, D.* C1 - 25985 C2 - 32006 SP - 3272-3291 TI - Rapamycin extends murine lifespan but has limited effects on aging. JO - J. Clin. Invest. VL - 123 IS - 3 PB - Amer. Soc. Clinical Investigation PY - 2013 SN - 0021-9738 ER - TY - JOUR AB - Autoreactive T cells can infiltrate the CNS to cause disorders such as multiple sclerosis. In order to visualize T cell activation in the CNS, we introduced a truncated fluorescent derivative of nuclear factor of activated T cells (NFAT) as a real-time T cell activation indicator. In experimental autoimmune encephalomyelitis, a rat model of multiple sclerosis, we tracked T cells interacting with structures of the vascular blood-brain barrier (BBB). 2-photon imaging documented the cytoplasmic-nuclear translocation of fluorescent NFAT, indicative of calcium-dependent activation of the T cells in the perivascular space, but not within the vascular lumen. The activation was related to contacts with the local antigen-presenting phagocytes and was noted only in T cells with a high pathogenic potential. T cell activation implied the presentation of an autoantigen, as the weakly pathogenic T cells, which remained silent in the untreated hosts, were activated upon instillation of exogenous autoantigen. Activation did not cogently signal long-lasting arrest, as individual T cells were able to sequentially contact fresh APCs. We propose that the presentation of local autoantigen by BBB-associated APCs provides stimuli that guide autoimmune T cells to the CNS destination, enabling them to attack the target tissue. AU - Pesic, M.* AU - Bartholomäus, I.* AU - Kyratsous, N.I.* AU - Heissmeyer, V. AU - Wekerle, H.* AU - Kawakami, N.* C1 - 23709 C2 - 31250 SP - 1192-1201 TI - 2-photon imaging of phagocyte-mediated T cell activation in the CNS. JO - J. Clin. Invest. VL - 123 IS - 3 PB - Amer. Soc. Clinical Investigation inc. PY - 2013 SN - 0021-9738 ER - TY - JOUR AB - Effector functions of inflammatory IL-17-producing Th (Th17) cells have been linked to autoimmune diseases such as experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis (MS). However, what determines Th17 cell encephalitogenicity is still unresolved. Here, we show that after EAE induction, mice deficient for the NF-κB regulator MALT1 (Malt1-/- mice) exhibit strong lymphocytic infiltration in the CNS, but do not develop any clinical signs of EAE. Loss of Malt1 interfered with expression of the Th17 effector cytokines IL-17 and GM-CSF both in vitro and in vivo. In line with their impaired GM-CSF secretion, Malt1-/- Th cells failed to recruit myeloid cells to the CNS to sustain neuroinflammation, whereas autoreactive WT Th cells successfully induced EAE in Malt1-/- hosts. In contrast, Malt1 deficiency did not affect Th1 cells. Despite their significantly decreased secretion of Th17 effector cytokines, Malt1-/- Th17 cells showed normal expression of lineage-specific transcription factors. Malt1-/- Th cells failed to cleave RelB, a suppressor of canonical NF-κB, and exhibited altered cellular localization of this protein. Our results indicate that MALT1 is a central, cell-intrinsic factor that determines the encephalitogenic potential of inflammatory Th17 cells in vivo. AU - Brüstle, A.* AU - Brenner, D.* AU - Knobbe, C.B.* AU - Lang, P.A.* AU - Virtanen, C.* AU - Hershenfield, B.M.* AU - Reardon, C.* AU - Lacher, S.M.* AU - Ruland, J. AU - Ohashi, P.S.* AU - Mak, T.W.* C1 - 11168 C2 - 30531 SP - 4698-4709 TI - The NF-κB regulator MALT1 determines the encephalitogenic potential of Th17 cells. JO - J. Clin. Invest. VL - 122 IS - 12 PB - American Society of Clinical Investigation Inc. PY - 2012 SN - 0021-9738 ER - TY - JOUR AB - The mutations that cause Leber congenital amaurosis (LCA) lead to photoreceptor cell death at an early age, causing childhood blindness. To unravel the molecular basis of LCA, we analyzed how mutations in LCA5 affect the connectivity of the encoded protein lebercilin at the interactome level. In photoreceptors, lebercilin is uniquely localized at the cilium that bridges the inner and outer segments. Using a generally applicable affinity proteomics approach, we showed that lebercilin specifically interacted with the intraflagellar transport (IFT) machinery in HEK293T cells. This interaction disappeared when 2 human LCA-associated lebercilin mutations were introduced, implicating a specific disruption of IFT-dependent protein transport, an evolutionarily conserved basic mechanism found in all cilia. Lca5 inactivation in mice led to partial displacement of opsins and light-induced translocation of arrestin from photoreceptor outer segments. This was consistent with a defect in IFT at the connecting cilium, leading to failure of proper outer segment formation and subsequent photoreceptor degeneration. These data suggest that lebercilin functions as an integral element of selective protein transport through photoreceptor cilia and provide a molecular demonstration that disrupted IFT can lead to LCA. AU - Boldt, K. AU - Mans, D.A.* AU - Won, J.* AU - van Reeuwijk, J.* AU - Vogt, A. AU - Kinkl, N. AU - Letteboer, S.J.* AU - Hicks, W.L.* AU - Hurd, R.E.* AU - Naggert, J.K.* AU - Texier, Y. AU - den Hollander, A.I.* AU - Koenekoop, R.K.* AU - Bennett, J.* AU - Cremers, F.P.* AU - Gloeckner, C.J. AU - Nishina, P.M.* AU - Roepman, R.* AU - Ueffing, M. C1 - 6424 C2 - 28648 SP - 2169-2180 TI - Disruption of intraflagellar protein transport in photoreceptor cilia causes Leber congenital amaurosis in humans and mice. JO - J. Clin. Invest. VL - 121 IS - 6 PB - Amer. Soc. Clinical Investigation Inc. PY - 2011 SN - 0021-9738 ER - TY - JOUR AB - Pulmonary emphysema is a disease characterized by alveolar cellular loss and inflammation. Recently, excessive apoptosis of structural alveolar cells has emerged as a major mechanism in the development of emphysema. Here, we investigated the proapoptotic and monocyte chemoattractant cytokine endothelial monocyte-activating protein 2 (EMAPII). Lung-specific overexpression of EMAPII in mice caused simplification of alveolar structures, apoptosis, and macrophage accumulation, compared with that in control transgenic mice. Additionally, in a mouse model of cigarette smoke-induced (CS-induced) emphysema, EMAPII levels were significantly increased in murine lungs. This upregulation was necessary for emphysema development, as neutralizing antibodies to EMAPII resulted in reduced alveolar cell apoptosis, inflammation, and emphysema-associated structural changes in alveoli and small airways and improved lung function. The mechanism of EMAPII upregulation involved an apoptosis-dependent feed-forward loop, since caspase-3 instillation in the lung markedly increased EMAPII expression, while caspase inhibition decreased its production, even in transgenic EMAPII mice. These findings may have clinical significance, as both current smokers and ex-smoker chronic obstructive pulmonary disease (COPD) patients had increased levels of secreted EMAPII in the bronchoalveolar lavage fluid compared with that of nonsmokers. In conclusion, we suggest that EMAPII perpetuates the mechanism of CS-induced lung emphysema in mice and, given its secretory nature, is a suitable target for neutralization antibody therapy. AU - Clauss, M.* AU - Voswinckel, R.* AU - Rajashekhar, G.* AU - Sigua, N.L.* AU - Fehrenbach, H.* AU - Rush, N.I.* AU - Schweitzer, K.S.* AU - Yildirim, A.Ö. AU - Kamocki, K.* AU - Fisher, A.J.* AU - Gu, Y.* AU - Safadi, B.* AU - Nikam, S.* AU - Hubbard, W.C.* AU - Tuder, R.M.* AU - Twigg, H.L.* AU - Presson, R.G.* AU - Sethi, S.* AU - Petrache, I.* C1 - 3949 C2 - 28687 SP - 2470-2479 TI - Lung endothelial monocyte-activating protein 2 is a mediator of cigarette smoke-induced emphysema in mice. JO - J. Clin. Invest. VL - 121 IS - 6 PB - Amer. Soc. Clinical Investigation Inc. PY - 2011 SN - 0021-9738 ER - TY - JOUR AB - Diarrheal diseases represent a major health burden in developing countries. Parenteral immunization typically does not induce efficient protection against enteropathogens because it does not stimulate migration of immune cells to the gut. Retinoic acid (RA) is critical for gut immunity, inducing upregulation of gut-homing receptors on activated T cells. In this study, we have demonstrated that RA can redirect immune responses elicited by s.c. vaccination of mice from skin-draining inguinal LNs (ingLNs) to the gut. When present during priming, RA induced robust upregulation of gut-homing receptors in ingLNs, imprinting gut-homing capacity on T cells. Concurrently, RA triggered the generation of gut-tropic IgA+ plasma cells in ingLNs and raised the levels of antigen-specific IgA in the intestinal lumen and blood. RA applied s.c. in vivo induced autonomous RA production in ingLN DCs, further driving efficient induction of gut-homing molecules on effector cells. Importantly, RA-supplemented s.c. immunization elicited a potent immune response in the small intestine that protected mice from cholera toxin-induced diarrhea and diminished bacterial loads in Peyer patches after oral infection with Salmonella. Thus, the use of RA as a gut-homing navigator represents a powerful tool to induce protective immunity in the intestine after s.c. immunization, offering what we believe to be a novel approach for vaccination against enteropathogens. AU - Hammerschmidt, S.I.* AU - Friedrichsen, M.* AU - Boelter, J.* AU - Lyszkiewicz, M.* AU - Kremmer, E. AU - Pabst, O.* AU - Forster, R.* C1 - 6426 C2 - 28657 SP - 3051-3061 TI - Retinoic acid induces homing of protective T and B cells to the gut after subcutaneous immunization in mice. JO - J. Clin. Invest. VL - 121 IS - 8 PB - American Soc. for Clinical Investigation PY - 2011 SN - 0021-9738 ER - TY - JOUR AB - Steroid-resistant nephrotic syndrome (SRNS) is a frequent cause of end-stage renal failure. Identification of single-gene causes of SRNS has generated some insights into its pathogenesis; however, additional genes and disease mechanisms remain obscure, and SRNS continues to be treatment refractory. Here we have identified 6 different mutations in coenzyme Q10 biosynthesis monooxygenase 6 (COQ6) in 13 individuals from 7 families by homozygosity mapping. Each mutation was linked to early-onset SRNS with sensorineural deafness. The deleterious effects of these human COQ6 mutations were validated by their lack of complementation in coq6-deficient yeast. Furthermore, knockdown of Coq6 in podocyte cell lines and coq6 in zebrafish embryos caused apoptosis that was partially reversed by coenzyme Q10 treatment. In rats, COQ6 was located within cell processes and the Golgi apparatus of renal glomerular podocytes and in stria vascularis cells of the inner ear, consistent with an oto-renal disease phenotype. These data suggest that coenzyme Q10-related forms of SRNS and hearing loss can be molecularly identified and potentially treated. AU - Heeringa, S.F.* AU - Chernin, G.* AU - Chaki, M.* AU - Zhou, W.* AU - Sloan, A.J.* AU - Ji, Z.* AU - Xie, L.X.* AU - Salviati, L.* AU - Hurd, T.W.* AU - Vega-Warner, V.* AU - Killen, P.D.* AU - Raphael, Y.* AU - Ashraf, S.* AU - Ovunc, B.* AU - Schoeb, D.S.* AU - McLaughlin, H.M.* AU - Airik, R.* AU - Vlangos, C.N.* AU - Gbadegesin, R.* AU - Hinkes, B.* AU - Saisawat, P.* AU - Trevisson, E.* AU - Doimo, M.* AU - Casarin, A.* AU - Pertegato, V.* AU - Giorgi, G.* AU - Prokisch, H. AU - Rotig, A.* AU - Nurnberg, G.* AU - Becker, C.* AU - Wang, S.* AU - Ozaltin, F.* AU - Topaloglu, R.* AU - Bakkaloglu, A.* AU - Bakkaloglu, S.A.* AU - Müller, D.* AU - Beissert, A.* AU - Mir, S.* AU - Berdeli, A.* AU - Varpizen, S.* AU - Zenker, M.* AU - Matejas, V.* AU - Santos-Ocaña, C.* AU - Navas, P.* AU - Kusakabe, T.* AU - Kispert, A.* AU - Akman, S.* AU - Soliman, N.A.* AU - Krick, S.* AU - Mundel, P.* AU - Reiser, J.* AU - Nürnberg, P.* AU - Clarke, C.F.* AU - Wiggins, R.C.* AU - Faul, C.* AU - Hildebrandt, F.* C1 - 5048 C2 - 28813 SP - 2013-2024 TI - COQ6 mutations in human patients produce nephrotic syndrome with sensorineural deafness. JO - J. Clin. Invest. VL - 121 IS - 5 PB - Amer Soc Clinical Investigation Inc. PY - 2011 SN - 0021-9738 ER - TY - JOUR AB - Wilson disease (WD) is a rare hereditary condition that is caused by a genetic defect in the copper-transporting ATPase ATP7B that results in hepatic copper accumulation and lethal liver failure. The present study focuses on the structural mitochondrial alterations that precede clinical symptoms in the livers of rats lacking Atp7b, an animal model for WD. Liver mitochondria from these Atp7b–/– rats contained enlarged cristae and widened intermembrane spaces, which coincided with a massive mitochondrial accumulation of copper. These changes, however, preceded detectable deficits in oxidative phosphorylation and biochemical signs of oxidative damage, suggesting that the ultrastructural modifications were not the result of oxidative stress imposed by copper- dependent Fenton chemistry. In a cell-free system containing a reducing dithiol agent, isolated mitochondria exposed to copper underwent modifications that were closely related to those observed in vivo. In this cell-free system, copper induced thiol modifications of three abundant mitochondrial membrane proteins, and this correlated with reversible intramitochondrial membrane crosslinking, which was also observed in liver mitochondria from Atp7b–/– rats. In vivo, copper-chelating agents reversed mitochondrial accumulation of copper, as well as signs of intra-mitochondrial membrane crosslinking, thereby preserving the functional and structural integrity of mitochondria. Together, these findings suggest that the mitochondrion constitutes a pivotal target of copper in WD. AU - Zischka, H. AU - Lichtmannegger, J. AU - Schmitt, S. AU - Jägemann, N. AU - Schulz, S. AU - Wartini, D. AU - Jennen, L. AU - Rust, C.* AU - Larochette, N.* AU - Galluzzi, L.* AU - Chajes, V.* AU - Bandow, N.* AU - Gilles, V.S.* AU - DiSpirito, A.A.* AU - Esposito, I.* AU - Göttlicher, M. AU - Summer, K.H. AU - Kroemer, G.* C1 - 6033 C2 - 29221 SP - 1508-1518 TI - Liver mitochondrial membrane crosslinking and destruction in a rat model of Wilson disease. JO - J. Clin. Invest. VL - 121 IS - 4 PB - Amer Soc Clinical Investigation Inc. PY - 2011 SN - 0021-9738 ER - TY - JOUR AB - The apoptosis inhibitor protein survivin is overexpressed in many tumors, making it a candidate target molecule for various forms of immunotherapy. To explore survivin as a target antigen for adoptive T cell therapy using lymphocytes expressing survivin-specific transgenic T cell receptors (Tg-TCRs), we isolated HLA-A2-allorestricted survivin-specific T cells with high functional avidity. Lymphocytes expressing Tg-TCRs were derived from these T cells and specifically recognized HLA-A2+ survivin+ tumor cells. Surprisingly, HLA-A2+ but not HLA-A2- lymphocytes expressing Tg-TCRs underwent extensive apoptosis over time. This demise was caused by HLA-A2-restricted fratricide that occurred due to survivin expression in lymphocytes, which created ligands for Tg-TCR recognition. Therefore, survivin-specific TCR gene therapy would be limited to application in HLA-A2-mismatched stem cell transplantation. We also noted that lymphocytes that expressed survivin-specific Tg-TCRs killed T cell clones of various specificities derived from HLA-A2+ but not HLA-A2- donors. These results raise a general question regarding the development of cancer vaccines that target proteins that are also expressed in activated lymphocytes, since induction of high-avidity T cells that expand in lymph nodes following vaccination or later accumulate at tumor sites might limit themselves by self-MHC-restricted fratricide while at the same time inadvertently eliminating neighboring T cells of other specificities. AU - Leisegang, M.* AU - Wilde, S. AU - Spranger, S. AU - Milosevic, S. AU - Frankenberger, B. AU - Uckert, W.* AU - Schendel, D.J. C1 - 5980 C2 - 27740 SP - 3869-3877 TI - MHC-restricted fratricide of human lymphocytes expressing survivin-specific transgenic T cell receptors. JO - J. Clin. Invest. VL - 120 IS - 11 PB - American Society for Clinical Investigation PY - 2010 SN - 0021-9738 ER - TY - JOUR AB - The autosomal recessive kidney disease nephronophthisis (NPHP) constitutes the most frequent genetic cause of terminal renal failure in the first 3 decades of life. Ten causative genes (NPHP1-NPHP9 and NPHP11), whose products localize to the primary cilia-centrosome complex, support the unifying concept that cystic kidney diseases are "ciliopathies". Using genome-wide homozygosity mapping, we report here what we believe to be a new locus (NPHP-like 1 [NPHPL1]) for an NPHP-like nephropathy. In 2 families with an NPHP-like phenotype, we detected homozygous frameshift and splice-site mutations, respectively, in the X-prolyl aminopeptidase 3 (XPNPEP3) gene. In contrast to all known NPHP proteins, XPNPEP3 localizes to mitochondria of renal cells. However, in vivo analyses also revealed a likely cilia-related function; suppression of zebrafish xpnpep3 phenocopied the developmental phenotypes of ciliopathy morphants, and this effect was rescued by human XPNPEP3 that was devoid of a mitochondrial localization signal. Consistent with a role for XPNPEP3 in ciliary function, several ciliary cystogenic proteins were found to be XPNPEP3 substrates, for which resistance to N-terminal proline cleavage resulted in attenuated protein function in vivo in zebrafish. Our data highlight an emerging link between mitochondria and ciliary dysfunction, and suggest that further understanding the enzymatic activity and substrates of XPNPEP3 will illuminate novel cystogenic pathways. AU - O'Toole, J.F.* AU - Liu, Y.J.* AU - Davis, E.E.* AU - Westlake, C.J.* AU - Attanasio, M.* AU - Otto, E.A.* AU - Seelow, D.* AU - Nurnberg, G.* AU - Becker, C.* AU - Nuutinen, M.* AU - Kärppa, M.* AU - Ignatius, J.* AU - Uusimaa, J.* AU - Pakanen, S.* AU - Jaakkola, E.* AU - van den Heuvel, L.P.* AU - Fehrenbach, H.* AU - Wiggins, R.* AU - Goyal, M.B.* AU - Zhou, W.* AU - Wolf, M.T.F.* AU - Wise, E.* AU - Helou, J.* AU - Allen, S.J.* AU - Murga-Zamalloa, C.A.* AU - Ashraf, S.* AU - Chaki, M.* AU - Heeringa, S.* AU - Chernin, G.* AU - Hoskins, B.E.* AU - Chaib, H.* AU - Gleeson, J.* AU - Kusakabe, T.* AU - Suzuki, T.* AU - Isaac, R.E.* AU - Quarmby, L.M.* AU - Tennant, B.* AU - Fujioka, H.* AU - Tuominen, H.* AU - Hassinen, I.* AU - Lohi, H.* AU - van Houten, J.L.* AU - Rotig, A.* AU - Sayer, J.A.* AU - Rolinski, B.* AU - Freisinger, P.* AU - Madhavan, S.M.* AU - Herzer, M. AU - Madignier, F. AU - Prokisch, H. AU - Nürnberg, P.* AU - Jackson, P.K.* AU - Khanna, H.* AU - Katsanis, N.* AU - Hildebrandt, F.* C1 - 2096 C2 - 27500 SP - 791-802 TI - Individuals with mutations in XPNPEP3, which encodes a mitochondrial protein, develop a nephronophthisis-like nephropathy. JO - J. Clin. Invest. VL - 120 IS - 3 PB - American Society Clinical Investigation Inc. PY - 2010 SN - 0021-9738 ER - TY - JOUR AB - Th subsets are defined according to their production of lineage-indicating cytokines and functions. In this study, we have identified a subset of human Th cells that infiltrates the epidermis in individuals with inflammatory skin disorders and is characterized by the secretion of IL-22 and TNF-alpha, but not IFN-gamma, IL-4, or IL-17. In analogy to the Th17 subset, cells with this cytokine profile have been named the Th22 subset. Th22 clones derived from patients with psoriasis were stable in culture and exhibited a transcriptome profile clearly separate from those of Th1, Th2, and Th17 cells; it included genes encoding proteins involved in tissue remodeling, such as FGFs, and chemokines involved in angiogenesis and fibrosis. Primary human keratinocytes exposed to Th22 supernatants expressed a transcriptome response profile that included genes involved in innate immune pathways and the induction and modulation of adaptive immunity. These proinflammatory Th22 responses were synergistically dependent on IL-22 and TNF-alpha. Furthermore, Th22 supernatants enhanced wound healing in an in vitro injury model, which was exclusively dependent on IL-22. In conclusion, the human Th22 subset may represent a separate T cell subset with a distinct identity with respect to gene expression and function, present within the epidermal layer in inflammatory skin diseases. Future strategies directed against the Th22 subset may be of value in chronic inflammatory skin disorders. AU - Eyerich, S.* AU - Eyerich, K.* AU - Pennino, D.* AU - Carbone, T.* AU - Nasorri, F.* AU - Pallotta, S.* AU - Cianfarani, F.* AU - Odorisio, T.* AU - Traidl-Hoffmann, C. AU - Behrendt, H. AU - Durham, S.R.* AU - Schmidt-Weber, C.B.* AU - Cavani, A.* C1 - 48567 C2 - 41188 SP - 3573-3585 TI - Th22 cells represent a distinct human T cell subset involved in epidermal immunity and remodeling. JO - J. Clin. Invest. VL - 119 IS - 12 PY - 2009 SN - 0021-9738 ER - TY - JOUR AB - The transcription factor serum response factor (SRF) plays a crucial role in the development of several organs. However, its role in the skin has not been explored. Here, we show that keratinocytes in normal human and mouse skin expressed high levels of SRF but that SRF expression was strongly downregulated in the hyperproliferative epidermis of wounded and psoriatic skin. Keratinocyte-specific deletion within the mouse SRF locus during embryonic development caused edema and skin blistering, and all animals died in utero. Postnatal loss of mouse SRF in keratinocytes resulted in the development of psoriasis-like skin lesions. These lesions were characterized by inflammation, hyperproliferation, and abnormal differentiation of keratinocytes as well as by disruption of the actin cytoskeleton. Ultrastructural analysis revealed markedly reduced cell-cell and cell-matrix contacts and loss of cell compaction in all epidermal layers. siRNA-mediated knockdown of SRF in primary human keratinocytes revealed that the cytoskeletal abnormalities and adhesion defects were a direct consequence of the loss of SRF. In contrast, the hyperproliferation observed in vivo was an indirect effect that was most likely a consequence of the inflammation. These results reveal that loss of SRF disrupts epidermal homeostasis and strongly suggest its involvement in the pathogenesis of hyperproliferative skin diseases, including psoriasis. AU - Koegel, H.* AU - von Tobel, L.* AU - Schäfer, M.* AU - Alberti, S.* AU - Kremmer, E. AU - Mauch, C.* AU - Hohl, D.* AU - Wang, X.J.* AU - Beer, H.-D.* AU - Bloch, W.* AU - Nordheim, A.* AU - Werner, S.* C1 - 997 C2 - 26514 SP - 899-910 TI - Loss of serum response factor in keratinocytes results in hyperproliferative skin disease in mice. JO - J. Clin. Invest. VL - 119 IS - 4 PB - Amer Soc Clinical Investigation Inc PY - 2009 SN - 0021-9738 ER - TY - JOUR AB - Idiopathic pulmonary fibrosis (IPF) is characterized by distorted lung architecture and loss of respiratory function. Enhanced (myo)fibroblast activation, ECM deposition, and alveolar epithelial type II (ATII) cell dysfunction contribute to IPF pathogenesis. However, the molecular pathways linking ATII cell dysfunction with the development of fibrosis are poorly understood. Here, we demonstrate, in a mouse model of pulmonary fibrosis, increased proliferation and altered expression of components of the WNT/beta-catenin signaling pathway in ATII cells. Further analysis revealed that expression of WNT1-inducible signaling protein-1 (WISP1), which is encoded by a WNT target gene, was increased in ATII cells in both a mouse model of pulmonary fibrosis and patients with IPF. Treatment of mouse primary ATII cells with recombinant WISP1 led to increased proliferation and epithelial-mesenchymal transition (EMT), while treatment of mouse and human lung fibroblasts with recombinant WISP1 enhanced deposition of ECM components. In the mouse model of pulmonary fibrosis, neutralizing mAbs specific for WISP1 reduced the expression of genes characteristic of fibrosis and reversed the expression of genes associated with EMT. More importantly, these changes in gene expression were associated with marked attenuation of lung fibrosis, including decreased collagen deposition and improved lung function and survival. Our study thus identifies WISP1 as a key regulator of ATII cell hyperplasia and plasticity as well as a potential therapeutic target for attenuation of pulmonary fibrosis. AU - Königshoff, M. AU - Kramer, M.* AU - Balsara, N.* AU - Wilhelm, J.* AU - Amarie, O.V.* AU - Jahn, A.* AU - Rose, F.* AU - Fink, L.* AU - Seeger, W.* AU - Schaefer, L.* AU - Günther, A.* AU - Eickelberg, O. C1 - 253 C2 - 26816 SP - 772-787 TI - WNT1-inducible signaling protein-1 mediates pulmonary fibrosis in mice and is upregulated in humans with idiopathic pulmonary fibrosis. JO - J. Clin. Invest. VL - 119 IS - 4 PB - American Society for Clinical Investigation PY - 2009 SN - 0021-9738 ER - TY - JOUR AB - Our aging society is confronted with a dramatic increase of patients suffering from tauopathies, which include Alzheimer disease and certain frontotemporal dementias. These disorders are characterized by typical neuropathological lesions including hyperphosphorylation and subsequent aggregation of TAU protein and neuronal cell death. Currently, no mechanism-based cures are available. We generated fluorescently labeled TAU transgenic zebrafish, which rapidly recapitulated key pathological features of tauopathies, including phosphorylation and conformational changes of human TAU protein, tangle formation, neuronal and behavioral disturbances, and cell death. Due to their optical transparency and small size, zebrafish larvae are well suited for both in vivo imaging and drug development. TAU-induced neuronal cell death was imaged by time-lapse microscopy in vivo. Furthermore, we used this zebrafish model to identify compounds targeting the TAU kinase glycogen synthase kinase 3(3 (GSK3 beta). We identified a newly developed highly active GSK3 beta inhibitor, AR-534, by rational drug design. AR-534 reduced TAU phosphorylation in TAU transgenic zebrafish. This transgenic zebrafish model may become a valuable tool for further studies of the neuropathology of dementia. AU - Paquet, D.* AU - Bhat, R.* AU - Sydow, A.* AU - Mandelkow, E.-M.* AU - Berg, S.* AU - Hellberg, S.* AU - Fälting, J.* AU - Distel, M. AU - Köster, R.W. AU - Schmid, B.* AU - Haass, C.* C1 - 1145 C2 - 26173 SP - 1382-1395 TI - A zebrafish model of tauopathy allows in vivo imaging of neuronal cell death and drug evaluation. JO - J. Clin. Invest. VL - 119 IS - 5 PB - Amer Soc Clinical Investigation Inc PY - 2009 SN - 0021-9738 ER - TY - JOUR AB - Neutrophil granulocytes form the body's first line of antibacterial defense, but they also contribute to tissue injury and noninfectious, chronic inflammation. Proteinase 3 (PR3) and neutrophil elastase (NE) are 2 abundant neutrophil serine proteases implicated in antimicrobial defense with overlapping and potentially redundant substrate specificity. Here, we unraveled a cooperative role for PR3 and NE in neutrophil activation and noninfectious inflammation in vivo, which we believe to be novel. Mice lacking both PR3 and NE demonstrated strongly diminished immune complex-mediated (IC-mediated) neutrophil infiltration in vivo as well as reduced activation of isolated neutrophils by ICs in vitro. In contrast, in mice lacking just NE, neutrophil recruitment to ICs was only marginally impaired. The defects in mice lacking both PR3 and NE were directly linked to the accumulation of antiinflammatory progranulin (PGRN). Both PR3 and NE cleaved PGRN in vitro and during neutrophil activation and inflammation in vivo. Local administration of recombinant PGRN potently inhibited neutrophilic inflammation in vivo, demonstrating that PGRN represents a crucial inflammation-suppressing mediator. We conclude that PR3 and NE enhance neutrophil-dependent inflammation by eliminating the local antiinflammatory activity of PGRN. Our results support the use of serine protease inhibitors as antiinflammatory agents. AU - Kessenbrock, K.* AU - Fröhlich, L.* AU - Sixt, M.* AU - Lämmermann, T.* AU - Pfister, H.* AU - Bateman, A.* AU - Belaaouaj, A.* AU - Ring, J. AU - Ollert, M.* AU - Fässler, R.* AU - Jenne, D.* C1 - 4079 C2 - 25776 SP - 2438-2447 TI - Proteinase 3 and neutrophil elastase enhance inflammation in mice by inactivating antiinflammatory progranulin. JO - J. Clin. Invest. VL - 118 IS - 7 PB - American Society for Clinical Investigation PY - 2008 SN - 0021-9738 ER - TY - JOUR AB - Brugada syndrome is a genetic disease associated with sudden cardiac death that is characterized by ventricular fibrillation and right precordial ST segment elevation on ECG. Loss-of-function mutations in SCN5A, which encodes the predominant cardiac sodium channel alpha subunit NaV1.5, can cause Brugada syndrome and cardiac conduction disease. However, SCN5A mutations are not detected in the majority of patients with these syndromes, suggesting that other genes can cause or modify presentation of these disorders. Here, we investigated SCN1B, which encodes the function-modifying sodium channel beta1 subunit, in 282 probands with Brugada syndrome and in 44 patients with conduction disease, none of whom had SCN5A mutations. We identified 3 mutations segregating with arrhythmia in 3 kindreds. Two of these mutations were located in a newly described alternately processed transcript, beta1B. Both the canonical and alternately processed transcripts were expressed in the human heart and were expressed to a greater degree in Purkinje fibers than in heart muscle, consistent with the clinical presentation of conduction disease. Sodium current was lower when NaV1.5 was coexpressed with mutant beta1 or beta1B subunits than when it was coexpressed with WT subunits. These findings implicate SCN1B as a disease gene for human arrhythmia susceptibility. AU - Watanabe, H.* AU - Koopmann, T.T.* AU - Le, Scouarnec, S.* AU - Yang, T.* AU - Ingram, C.R.* AU - Schott, J.J.* AU - Demolombe, S.* AU - Probst, V.* AU - Anselme, F.* AU - Escande, D.* AU - Wiesfeld, A.C.* AU - Pfeufer, A. AU - Kääb, S.* AU - Wichmann, H.-E. AU - Hasdemir, C.* AU - Aizawa, Y.* AU - Wilde, A.A.* AU - Roden, D.M.* AU - Bezzina, C.R.* C1 - 2495 C2 - 25495 SP - 2260-2268 TI - Sodium channel β1 subunit mutations associated with Brugada syndrome and cardiac conduction disease in humans. JO - J. Clin. Invest. VL - 118 IS - 6 PB - American Society for Clinical Investigation PY - 2008 SN - 0021-9738 ER - TY - JOUR AB - The molecular characterization of leukemia has demonstrated that genetic alterations in the leukemic clone frequently fall into 2 classes, those affecting transcription factors (e.g., AML1-ETO) and mutations affecting genes involved in signal transduction (e.g., activating mutations of FLT3 and KIT). This finding has favored a model of leukemogenesis in which the collaboration of these 2 classes of genetic alterations is necessary for the malignant transformation of hematopoietic progenitor cells. The model is supported by experimental data indicating that AML1-ETO and FLT3 length mutation (FLT3-LM), 2 of the most frequent genetic alterations in AML, are both insufficient on their own to cause leukemia in animal models. Here we report that AML1-ETO collaborates with FLT3-LM in inducing acute leukemia in a murine BM transplantation model. Moreover, in a series of 135 patients with AML1-ETO-positive AML, the most frequently identified class of additional mutations affected genes involved in signal transduction pathways including FLT3-LM or mutations of KIT and NRAS. These data support the concept of oncogenic cooperation between AML1-ETO and a class of activating mutations, recurrently found in patients with t(8;21), and provide a rationale for therapies targeting signal transduction pathways in AML1-ETO-positive leukemias. AU - Schessl, C. AU - Rawat, V.P.S. AU - Cusan, M. AU - Deshpande, A. AU - Kohl, T.M. AU - Spiekermann, K. AU - Hiddemann, W. AU - Quintanilla-Martinez, L. AU - Bohlander, S.K. AU - Feuring-Buske, M. AU - Buske, C. C1 - 5123 C2 - 22746 SP - 2159-2186 TI - The AML1-ETO fusion gene and the FLT3 lenght mutation collaborate in inducing acute leukemia in mice. JO - J. Clin. Invest. VL - 115 IS - 8 PY - 2005 SN - 0021-9738 ER -