62 results on '"Berriel Diaz M"'
Search Results
2. Mind the Ga(b)p! – A novel hepatic gatekeeper at the switch point of metabolic homeostasis and diabetic late complications controlled by reactive metabolites
- Author
-
Niopek, K, primary, Berriel Diaz, M, additional, Nawroth, P, additional, and Herzig, S, additional
- Published
- 2015
- Full Text
- View/download PDF
3. Transforming growth factor beta-like stimulated clone 22 D4 promotes diabetic hyperglycemia and insulin resistance
- Author
-
Friedrich, K, primary, Ekim Üstünel, B, additional, Wang, X, additional, Jones, A, additional, Rohm, M, additional, Berriel Diaz, M, additional, Stremmel, W, additional, Blüher, M, additional, and Herzig, S, additional
- Published
- 2015
- Full Text
- View/download PDF
4. P0413 : D-dopachrome tautomerase: Novel regulator of hepatic autophagy
- Author
-
Eheim, A.L., primary, Bahr, C., additional, Berriel Diaz, M., additional, and Herzig, S., additional
- Published
- 2015
- Full Text
- View/download PDF
5. P53 METABOLIC AND ONCOGENIC CONTRIBUTIONS OF MHC-AP IN OBESITY-ASSOCIATED HEPATOCELLULAR CARCINOMA
- Author
-
Eheim, A.L., primary, Berriel Diaz, M., additional, Meissburger, B., additional, and Herzig, S., additional
- Published
- 2014
- Full Text
- View/download PDF
6. The Glucocorticoid Receptor Controls Hepatic Dyslipidemia through Hes1
- Author
-
Lemke, U., Krones-Herzig, A., Berriel Diaz, M., Narvekar, P., Ziegler, A., Vegiopoulos, A., Cato, A.C.B., Bohl, S., Klingmüller, U., Screaton, R.A., Müller-Decker, K., Kersten, A.H., Herzig, S., Lemke, U., Krones-Herzig, A., Berriel Diaz, M., Narvekar, P., Ziegler, A., Vegiopoulos, A., Cato, A.C.B., Bohl, S., Klingmüller, U., Screaton, R.A., Müller-Decker, K., Kersten, A.H., and Herzig, S.
- Abstract
Aberrant accumulation of lipids in the liver (¿fatty liver¿ or hepatic steatosis) represents a hallmark of the metabolic syndrome and is tightly associated with obesity, type II diabetes, starvation, or glucocorticoid (GC) therapy. While fatty liver has been connected with numerous abnormalities of liver function, the molecular mechanisms of fatty liver development remain largely enigmatic. Here we show that liver-specific disruption of glucocorticoid receptor (GR) action improves the steatotic phenotype in fatty liver mouse models and leads to the induction of transcriptional repressor hairy enhancer of split 1 (Hes1) gene expression. The GR directly interferes with Hes1 promoter activity, triggering the recruitment of histone deacetylase (HDAC) activities to the Hes1 gene. Genetic restoration of hepatic Hes1 levels in steatotic animals normalizes hepatic triglyceride (TG) levels. As glucocorticoid action is increased during starvation, myotonic dystrophy, and Cushing's syndrome, the inhibition of Hes1 through the GR might explain the fatty liver phenotype in these subjects.
- Published
- 2008
7. Transforming growth factor beta 1-stimulated clone (TSC) 22 D4 is a novel key transcriptional co-regulator of hepatic metabolism
- Author
-
Friedrich, K, primary, Stremmel, W, additional, Berriel-Diaz, M, additional, and Herzig, S, additional
- Published
- 2013
- Full Text
- View/download PDF
8. Seasonal changes of myostatin expression and its relation to body Mass acclimation in the Djungarian hamster, Phodopus sungorus
- Author
-
Braulke, L.J., primary, Heldmaier, G., additional, Berriel Diaz, M., additional, Rozman, J., additional, and Exner, C., additional
- Published
- 2010
- Full Text
- View/download PDF
9. Control of hepatic and systemic lipid metabolism through the nuclear receptor co-factor RIP140
- Author
-
Berriel Diaz, M, primary, Krones-Herzig, A, additional, Ziegler, A, additional, Metzger, D, additional, and Herzig, S, additional
- Published
- 2007
- Full Text
- View/download PDF
10. Control of hepatic gluconeogenesis through nuclear receptor co-factor RIP140
- Author
-
Krones-Herzig, A, primary, Berriel Diaz, M, additional, Metzger, D, additional, Ziegler, A, additional, and Herzig, S, additional
- Published
- 2006
- Full Text
- View/download PDF
11. Control of macrophage TNFalpha expression through transcriptional co-factor RIP140
- Author
-
Zschiedrich, I, primary, Rieser, N, additional, Berriel Diaz, M, additional, and Herzig, S, additional
- Published
- 2006
- Full Text
- View/download PDF
12. Seasonal Changes of Myostatin Expression and Its Relation to Body Mass Acclimation in the Djungarian Hamster, Phodopus sungorus.
- Author
-
Braulkei, L. J., Heldmaier, G., Berriel Diaz, M., Rozman, J., and Exner, C.
- Subjects
HAMSTERS as laboratory animals ,ACCLIMATIZATION ,LEAN body mass ,BODY mass index ,ZOOLOGICAL research ,PHOTOPERIODISM - Abstract
The article presents a study which examines the seasonal changes of myostatin and its association to body mass in a Djungarian hamster in Hamburg, Germany. A total of 128 Djungarian hamsters, Phodopus sungorus, were bred and raised at the University of Marburg under natural photoperiod (NP). Findings of the study suggest that myostatin is seasonally regulated and contributes to low muscle mass (MM) and hence low lean mass (LM) and body mass (BM) during seasonal acclimatization in the Djungarian hamster.
- Published
- 2010
- Full Text
- View/download PDF
13. Virtual reality-empowered deep-learning analysis of brain cells.
- Author
-
Kaltenecker D, Al-Maskari R, Negwer M, Hoeher L, Kofler F, Zhao S, Todorov M, Rong Z, Paetzold JC, Wiestler B, Piraud M, Rueckert D, Geppert J, Morigny P, Rohm M, Menze BH, Herzig S, Berriel Diaz M, and Ertürk A
- Subjects
- Animals, Mice, Neurons, Software, Image Processing, Computer-Assisted methods, Proto-Oncogene Proteins c-fos metabolism, Humans, Deep Learning, Brain diagnostic imaging, Virtual Reality
- Abstract
Automated detection of specific cells in three-dimensional datasets such as whole-brain light-sheet image stacks is challenging. Here, we present DELiVR, a virtual reality-trained deep-learning pipeline for detecting c-Fos
+ cells as markers for neuronal activity in cleared mouse brains. Virtual reality annotation substantially accelerated training data generation, enabling DELiVR to outperform state-of-the-art cell-segmenting approaches. Our pipeline is available in a user-friendly Docker container that runs with a standalone Fiji plugin. DELiVR features a comprehensive toolkit for data visualization and can be customized to other cell types of interest, as we did here for microglia somata, using Fiji for dataset-specific training. We applied DELiVR to investigate cancer-related brain activity, unveiling an activation pattern that distinguishes weight-stable cancer from cancers associated with weight loss. Overall, DELiVR is a robust deep-learning tool that does not require advanced coding skills to analyze whole-brain imaging data in health and disease., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
14. Endothelial Notch1 signaling in white adipose tissue promotes cancer cachexia.
- Author
-
Taylor J, Uhl L, Moll I, Hasan SS, Wiedmann L, Morgenstern J, Giaimo BD, Friedrich T, Alsina-Sanchis E, De Angelis Rigotti F, Mülfarth R, Kaltenbach S, Schenk D, Nickel F, Fleming T, Sprinzak D, Mogler C, Korff T, Billeter AT, Müller-Stich BP, Berriel Diaz M, Borggrefe T, Herzig S, Rohm M, Rodriguez-Vita J, and Fischer A
- Subjects
- Animals, Humans, Male, Mice, Signal Transduction, Tretinoin, Adipose Tissue, White pathology, Cachexia pathology, Neoplasms complications, Receptor, Notch1 metabolism
- Abstract
Cachexia is a major cause of morbidity and mortality in individuals with cancer and is characterized by weight loss due to adipose and muscle tissue wasting. Hallmarks of white adipose tissue (WAT) remodeling, which often precedes weight loss, are impaired lipid storage, inflammation and eventually fibrosis. Tissue wasting occurs in response to tumor-secreted factors. Considering that the continuous endothelium in WAT is the first line of contact with circulating factors, we postulated whether the endothelium itself may orchestrate tissue remodeling. Here, we show using human and mouse cancer models that during precachexia, tumors overactivate Notch1 signaling in distant WAT endothelium. Sustained endothelial Notch1 signaling induces a WAT wasting phenotype in male mice through excessive retinoic acid production. Pharmacological blockade of retinoic acid signaling was sufficient to inhibit WAT wasting in a mouse cancer cachexia model. This demonstrates that cancer manipulates the endothelium at distant sites to mediate WAT wasting by altering angiocrine signals., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
15. Fasting-sensitive SUMO-switch on Prox1 controls hepatic cholesterol metabolism.
- Author
-
Alfaro AJ, Dittner C, Becker J, Loft A, Mhamane A, Maida A, Georgiadi A, Tsokanos FF, Klepac K, Molocea CE, El-Merahbi R, Motzler K, Geppert J, Karikari RA, Szendrödi J, Feuchtinger A, Hofmann S, Karaca S, Urlaub H, Berriel Diaz M, Melchior F, and Herzig S
- Abstract
Accumulation of excess nutrients hampers proper liver function and is linked to nonalcoholic fatty liver disease (NAFLD) in obesity. However, the signals responsible for an impaired adaptation of hepatocytes to obesogenic dietary cues remain still largely unknown. Post-translational modification by the small ubiquitin-like modifier (SUMO) allows for a dynamic regulation of numerous processes including transcriptional reprogramming. We demonstrate that specific SUMOylation of transcription factor Prox1 represents a nutrient-sensitive determinant of hepatic fasting metabolism. Prox1 is highly SUMOylated on lysine 556 in the liver of ad libitum and refed mice, while this modification is abolished upon fasting. In the context of diet-induced obesity, Prox1 SUMOylation becomes less sensitive to fasting cues. The hepatocyte-selective knock-in of a SUMOylation-deficient Prox1 mutant into mice fed a high-fat/high-fructose diet leads to a reduction of systemic cholesterol levels, associated with the induction of liver bile acid detoxifying pathways during fasting. The generation of tools to maintain the nutrient-sensitive SUMO-switch on Prox1 may thus contribute to the development of "fasting-based" approaches for the preservation of metabolic health., (© 2023 The Authors. Published under the terms of the CC BY NC ND 4.0 license.)
- Published
- 2023
- Full Text
- View/download PDF
16. Development of a peptide drug restoring AMPK and adipose tissue functionality in cancer cachexia.
- Author
-
Ji H, Englmaier F, Morigny P, Giroud M, Gräsle P, Brings S, Szendrödi J, Berriel Diaz M, Plettenburg O, Herzig S, and Rohm M
- Subjects
- Animals, Humans, Adipose Tissue metabolism, Cachexia drug therapy, Cachexia etiology, Cachexia metabolism, Peptides pharmacology, Pharmaceutical Preparations metabolism, Quality of Life, AMP-Activated Protein Kinases metabolism, Neoplasms complications, Neoplasms metabolism
- Abstract
Cancer cachexia is a severe systemic wasting disease that negatively affects quality of life and survival in patients with cancer. To date, treating cancer cachexia is still a major unmet clinical need. We recently discovered the destabilization of the AMP-activated protein kinase (AMPK) complex in adipose tissue as a key event in cachexia-related adipose tissue dysfunction and developed an adeno-associated virus (AAV)-based approach to prevent AMPK degradation and prolong cachexia-free survival. Here, we show the development and optimization of a prototypic peptide, Pen-X-ACIP, where the AMPK-stabilizing peptide ACIP is fused to the cell-penetrating peptide moiety penetratin via a propargylic glycine linker to enable late-stage functionalization using click chemistry. Pen-X-ACIP was efficiently taken up by adipocytes, inhibited lipolysis, and restored AMPK signaling. Tissue uptake assays showed a favorable uptake profile into adipose tissue upon intraperitoneal injection. Systemic delivery of Pen-X-ACIP into tumor-bearing animals prevented the progression of cancer cachexia without affecting tumor growth and preserved body weight and adipose tissue mass with no discernable side effects in other peripheral organs, thereby achieving proof of concept. As Pen-X-ACIP also exerted its anti-lipolytic activity in human adipocytes, it now provides a promising platform for further (pre)clinical development toward a novel, first-in-class approach against cancer cachexia., Competing Interests: Declaration of interests This work was in part supported by research grants by Novo Nordisk to O.P., S.H., and M.R., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
17. Trip13 Depletion in Liver Cancer Induces a Lipogenic Response Contributing to Plin2-Dependent Mitotic Cell Death.
- Author
-
Rios Garcia M, Meissburger B, Chan J, de Guia RM, Mattijssen F, Roessler S, Birkenfeld AL, Raschzok N, Riols F, Tokarz J, Giroud M, Gil Lozano M, Hartleben G, Nawroth P, Haid M, López M, Herzig S, and Berriel Diaz M
- Subjects
- ATPases Associated with Diverse Cellular Activities metabolism, Cell Cycle Proteins metabolism, Cell Death, Humans, Lipids, Mad2 Proteins metabolism, Paclitaxel pharmacology, Perilipin-2, Proto-Oncogene Proteins c-akt metabolism, Receptors, Thyroid Hormone metabolism, Insulins metabolism, Liver Neoplasms
- Abstract
Aberrant energy metabolism and cell cycle regulation both critically contribute to malignant cell growth and both processes represent targets for anticancer therapy. It is shown here that depletion of the AAA+-ATPase thyroid hormone receptor interacting protein 13 (Trip13) results in mitotic cell death through a combined mechanism linking lipid metabolism to aberrant mitosis. Diminished Trip13 levels in hepatocellular carcinoma cells result in insulin-receptor-/Akt-pathway-dependent accumulation of lipid droplets, which act as functional acentriolar microtubule organizing centers disturbing mitotic spindle polarity. Specifically, the lipid-droplet-coating protein perilipin 2 (Plin2) is required for multipolar spindle formation, induction of DNA damage, and mitotic cell death. Plin2 expression in different tumor cells confers susceptibility to cell death induced by Trip13 depletion as well as treatment with paclitaxel, a spindle-interfering drug commonly used against different cancers. Thus, assessment of Plin2 levels enables the stratification of tumor responsiveness to mitosis-targeting drugs, including clinically approved paclitaxel and Trip13 inhibitors currently under development., (© 2022 The Authors. Advanced Science published by Wiley-VCH GmbH.)
- Published
- 2022
- Full Text
- View/download PDF
18. Obesity and cancer-extracellular matrix, angiogenesis, and adrenergic signaling as unusual suspects linking the two diseases.
- Author
-
Pellegata NS, Berriel Diaz M, Rohm M, and Herzig S
- Subjects
- Adipose Tissue, Extracellular Matrix, Humans, Obesity complications, Adrenergic Agents, Neoplasms epidemiology
- Abstract
Obesity is an established risk factor for several human cancers. Given the association between excess body weight and cancer, the increasing rates of obesity worldwide are worrisome. A variety of obesity-related factors has been implicated in cancer initiation, progression, and response to therapy. These factors include circulating nutritional factors, hormones, and cytokines, causing hyperinsulinemia, inflammation, and adipose tissue dysfunction. The impact of these conditions on cancer development and progression has been the focus of extensive literature. In this review, we concentrate on processes that can link obesity and cancer, and which provide a novel perspective: extracellular matrix remodeling, angiogenesis, and adrenergic signaling. We describe molecular mechanisms involved in these processes, which represent putative targets for intervention. Liver, pancreas, and breast cancers were chosen as exemplary disease models. In view of the expanding epidemic of obesity, a better understanding of the tumorigenic process in obese individuals might lead to more effective treatments and preventive measures., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
19. Therapy-Related Transcriptional Subtypes in Matched Primary and Recurrent Head and Neck Cancer.
- Author
-
Weber P, Künstner A, Hess J, Unger K, Marschner S, Idel C, Ribbat-Idel J, Baumeister P, Gires O, Walz C, Rietzler S, Valeanu L, Herkommer T, Kreutzer L, Klymenko O, Drexler G, Kirchner T, Maihöfer C, Ganswindt U, Walch A, Sterr M, Lickert H, Canis M, Rades D, Perner S, Berriel Diaz M, Herzig S, Lauber K, Wollenberg B, Busch H, Belka C, and Zitzelsberger H
- Subjects
- Humans, Neoplasm Recurrence, Local genetics, RNA, Squamous Cell Carcinoma of Head and Neck genetics, Carcinoma, Squamous Cell genetics, Carcinoma, Squamous Cell pathology, Head and Neck Neoplasms genetics
- Abstract
Purpose: The genetic relatedness between primary and recurrent head and neck squamous cell carcinomas (HNSCC) reflects the extent of heterogeneity and therapy-driven selection of tumor subpopulations. Yet, current treatment of recurrent HNSCC ignores the molecular characteristics of therapy-resistant tumor populations., Experimental Design: From 150 tumors, 74 primary HNSCCs were RNA sequenced and 38 matched primary/recurrent tumor pairs were both whole-exome and RNA sequenced. Transcriptome analysis determined the predominant classical (CL), basal (BA), and inflamed-mesenchymal (IMS) transcriptional subtypes according to an established classification. Genomic alterations and clonal compositions of tumors were evaluated from whole-exome data., Results: Although CL and IMS subtypes were more common in primary HNSCC with low recurrence rates, the BA subtype was more prevalent and stable in recurrent tumors. The BA subtype was associated with a transcriptional signature of partial epithelial-to-mesenchymal transition (p-EMT) and early recurrence. In 44% of matched cases, the dominant subtype changed from primary to recurrent tumors, preferably from IMS to BA or CL. Expression analysis of prognostic gene sets identified upregulation of hypoxia, p-emt, and radiotherapy resistance signatures and downregulation of tumor inflammation in recurrences compared with index tumors. A relevant subset of primary/recurrent tumor pairs presented no evidence for a common clonal origin., Conclusions: Our study showed a high degree of genetic and transcriptional heterogeneity between primary/recurrent tumors, suggesting therapy-related selection of a transcriptional subtype with characteristics unfavorable for therapy. We conclude that therapy decisions should be based on genetic and transcriptional characteristics of recurrences rather than primary tumors to enable optimally tailored treatment strategies., (©2021 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
20. Aging Aggravates Cachexia in Tumor-Bearing Mice.
- Author
-
Geppert J, Walth AA, Terrón Expósito R, Kaltenecker D, Morigny P, Machado J, Becker M, Simoes E, Lima JDCC, Daniel C, Berriel Diaz M, Herzig S, Seelaender M, and Rohm M
- Abstract
Background: Cancer is primarily a disease of high age in humans, yet most mouse studies on cancer cachexia are conducted using young adolescent mice. Given that metabolism and muscle function change with age, we hypothesized that aging may affect cachexia progression in mouse models., Methods: We compare tumor and cachexia development in young and old mice of three different strains (C57BL/6J, C57BL/6N, BALB/c) and with two different tumor cell lines (Lewis Lung Cancer, Colon26). Tumor size, body and organ weights, fiber cross-sectional area, circulating cachexia biomarkers, and molecular markers of muscle atrophy and adipose tissue wasting are shown. We correlate inflammatory markers and body weight dependent on age in patients with cancer., Results: We note fundamental differences between mouse strains. Aging aggravates weight loss in LLC-injected C57BL/6J mice, drives it in C57BL/6N mice, and does not influence weight loss in C26-injected BALB/c mice. Glucose tolerance is unchanged in cachectic young and old mice. The stress marker GDF15 is elevated in cachectic BALB/c mice independent of age and increased in old C57BL/6N and J mice. Inflammatory markers correlate significantly with weight loss only in young mice and patients., Conclusions: Aging affects cachexia development and progression in mice in a strain-dependent manner and influences the inflammatory profile in both mice and patients. Age is an important factor to consider for future cachexia studies.
- Published
- 2021
- Full Text
- View/download PDF
21. Association of circulating PLA2G7 levels with cancer cachexia and assessment of darapladib as a therapy.
- Author
-
Morigny P, Kaltenecker D, Zuber J, Machado J, Mehr L, Tsokanos FF, Kuzi H, Hermann CD, Voelkl M, Monogarov G, Springfeld C, Laurent V, Engelmann B, Friess H, Zörnig I, Krüger A, Krijgsveld J, Prokopchuk O, Fisker Schmidt S, Rohm M, Herzig S, and Berriel Diaz M
- Subjects
- 1-Alkyl-2-acetylglycerophosphocholine Esterase, Animals, Benzaldehydes, Biomarkers, Humans, Mice, Oximes, Prospective Studies, Cachexia drug therapy, Cachexia etiology, Pancreatic Neoplasms
- Abstract
Background: Cancer cachexia (CCx) is a multifactorial wasting disorder characterized by involuntary loss of body weight that affects many cancer patients and implies a poor prognosis, reducing both tolerance to and efficiency of anticancer therapies. Actual challenges in management of CCx remain in the identification of tumour-derived and host-derived mediators involved in systemic inflammation and tissue wasting and in the discovery of biomarkers that would allow for an earlier and personalized care of cancer patients. The aim of this study was to identify new markers of CCx across different species and tumour entities., Methods: Quantitative secretome analysis was performed to identify specific factors characteristic of cachexia-inducing cancer cell lines. To establish the subsequently identified phospholipase PLA2G7 as a marker of CCx, plasma PLA2G7 activity and/or protein levels were measured in well-established mouse models of CCx and in different cohorts of weight-stable and weight-losing cancer patients with different tumour entities. Genetic PLA2G7 knock-down in tumours and pharmacological treatment using the well-studied PLA2G7 inhibitor darapladib were performed to assess its implication in the pathogenesis of CCx in C26 tumour-bearing mice., Results: High expression and secretion of PLA2G7 were hallmarks of cachexia-inducing cancer cell lines. Circulating PLA2G7 activity was increased in different mouse models of CCx with various tumour entities and was associated with the severity of body wasting. Circulating PLA2G7 levels gradually rose during cachexia development. Genetic PLA2G7 knock-down in C26 tumours only partially reduced plasma PLA2G7 levels, suggesting that the host is also an important contributor. Chronic treatment with darapladib was not sufficient to counteract inflammation and tissue wasting despite a strong inhibition of the circulating PLA2G7 activity. Importantly, PLA2G7 levels were also increased in colorectal and pancreatic cancer patients with CCx., Conclusions: Overall, our data show that despite no immediate pathogenic role, at least when targeted as a single entity, PLA2G7 is a consistent marker of CCx in both mice and humans. The early increase in circulating PLA2G7 levels in pre-cachectic mice supports future prospective studies to assess its potential as biomarker for cancer patients., (© 2021 The Authors. Journal of Cachexia, Sarcopenia and Muscle published by John Wiley & Sons Ltd on behalf of Society on Sarcopenia, Cachexia and Wasting Disorders.)
- Published
- 2021
- Full Text
- View/download PDF
22. Imaging modalities for diagnosis and monitoring of cancer cachexia.
- Author
-
Han J, Harrison L, Patzelt L, Wu M, Junker D, Herzig S, Berriel Diaz M, and Karampinos DC
- Abstract
Cachexia, a multifactorial wasting syndrome, is highly prevalent among advanced-stage cancer patients. Unlike weight loss in healthy humans, the progressive loss of body weight in cancer cachexia primarily implicates lean body mass, caused by an aberrant metabolism and systemic inflammation. This may lead to disease aggravation, poorer quality of life, and increased mortality. Timely detection is, therefore, crucial, as is the careful monitoring of cancer progression, in an effort to improve management, facilitate individual treatment and minimize disease complications. A detailed analysis of body composition and tissue changes using imaging modalities-that is, computed tomography, magnetic resonance imaging, (
18 F) fluoro-2-deoxy-D-glucose (18 FDG) PET and dual-energy X-ray absorptiometry-shows great premise for charting the course of cachexia. Quantitative and qualitative changes to adipose tissue, organs, and muscle compartments, particularly of the trunk and extremities, could present important biomarkers for phenotyping cachexia and determining its onset in patients. In this review, we present and compare the imaging techniques that have been used in the setting of cancer cachexia. Their individual limitations, drawbacks in the face of clinical routine care, and relevance in oncology are also discussed., (© 2021. The Author(s).)- Published
- 2021
- Full Text
- View/download PDF
23. Combination therapies induce cancer cell death through the integrated stress response and disturbed pyrimidine metabolism.
- Author
-
Hartleben G, Schorpp K, Kwon Y, Betz B, Tsokanos FF, Dantes Z, Schäfer A, Rothenaigner I, Monroy Kuhn JM, Morigny P, Mehr L, Lin S, Seitz S, Tokarz J, Artati A, Adamsky J, Plettenburg O, Lutter D, Irmler M, Beckers J, Reichert M, Hadian K, Zeigerer A, Herzig S, and Berriel Diaz M
- Subjects
- Cell Death, Humans, Niclosamide, Pyrimidines, Antineoplastic Agents, Neoplasms
- Abstract
By accentuating drug efficacy and impeding resistance mechanisms, combinatorial, multi-agent therapies have emerged as key approaches in the treatment of complex diseases, most notably cancer. Using high-throughput drug screens, we uncovered distinct metabolic vulnerabilities and thereby identified drug combinations synergistically causing a starvation-like lethal catabolic response in tumor cells from different cancer entities. Domperidone, a dopamine receptor antagonist, as well as several tricyclic antidepressants (TCAs), including imipramine, induced cancer cell death in combination with the mitochondrial uncoupler niclosamide ethanolamine (NEN) through activation of the integrated stress response pathway and the catabolic CLEAR network. Using transcriptome and metabolome analyses, we characterized a combinatorial response, mainly driven by the transcription factors CHOP and TFE3, which resulted in cell death through enhanced pyrimidine catabolism as well as reduced pyrimidine synthesis. Remarkably, the drug combinations sensitized human organoid cultures to the standard-of-care chemotherapy paclitaxel. Thus, our combinatorial approach could be clinically implemented into established treatment regimen, which would be further facilitated by the advantages of drug repurposing., (© 2021 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2021
- Full Text
- View/download PDF
24. High levels of modified ceramides are a defining feature of murine and human cancer cachexia.
- Author
-
Morigny P, Zuber J, Haid M, Kaltenecker D, Riols F, Lima JDC, Simoes E, Otoch JP, Schmidt SF, Herzig S, Adamski J, Seelaender M, Berriel Diaz M, and Rohm M
- Subjects
- Animals, Humans, Mice, Muscular Atrophy, Quality of Life, Cachexia etiology, Ceramides metabolism, Neoplasms complications
- Abstract
Background: Cancer cachexia (CCx) is a multifactorial energy-wasting syndrome reducing the efficiency of anti-cancer therapies, quality of life, and survival of cancer patients. In the past years, most studies focused on the identification of tumour and host-derived proteins contributing to CCx. However, there is still a lack of studies addressing the changes in bioactive lipids. The aim of this study was to identify specific lipid species as a hallmark of CCx by performing a broad range lipid analysis of plasma from well-established CCx mouse models as well as cachectic and weight stable cancer patients., Methods: Plasma from non-cachectic (PBS-injected mice, NC26 tumour-bearing mice), pre-cachectic and cachectic mice (C26 and LLC tumour-bearing mice, Apc
Min/+ mutant mice), and plasma from weight stable and cachectic patients with gastrointestinal cancer, were analysed using the Lipidyzer™ platform. In total, 13 lipid classes and more than 1100 lipid species, including sphingolipids, neutral and polar glycerolipids, were covered by the analysis. Correlation analysis between specific lipid species and readouts of CCx were performed. Lipidomics data were confirmed by gene expression analysis of metabolic organs to analyse enzymes involved in sphingolipid synthesis and degradation., Results: A decrease in several lysophosphatidylcholine (LPC) species and an increase in numerous sphingolipids including sphingomyelins (SMs), ceramides (CERs), hexosyl-ceramides (HCERs) and lactosyl-ceramides (LCERs), were mutual features of CCx in both mice and cancer patients. Notably, sphingolipid levels gradually increased during cachexia development. Key enzymes involved in ceramide synthesis were elevated in liver but not in adipose, muscle, or tumour tissues, suggesting that ceramide turnover in the liver is a major contributor to elevated sphingolipid levels in CCx. LPC(16:1), LPC(20:3), SM(16:0), SM(24:1), CER(16:0), CER(24:1), HCER(16:0), and HCER(24:1) were the most consistently affected lipid species between mice and humans and correlated negatively (LPCs) or positively (SMs, CERs and HCERs) with the severity of body weight loss., Conclusions: High levels of sphingolipids, specifically ceramides and modified ceramides, are a defining feature of murine and human CCx and may contribute to tissue wasting and skeletal muscle atrophy through the inhibition of anabolic signals. The progressive increase in sphingolipids during cachexia development supports their potential as early biomarkers for CCx., (© 2020 The Authors. Journal of Cachexia, Sarcopenia and Muscle published by John Wiley & Sons Ltd on behalf of Society on Sarcopenia, Cachexia and Wasting Disorders.)- Published
- 2020
- Full Text
- View/download PDF
25. 'Corrigendum to "Ataxin-10 is part of a cachexokine cocktail triggering cardiac metabolic dysfunction in cancer cachexia" [Molecular Metabolism 5 (2) (2015) 67-78]'.
- Author
-
Schäfer M, Oeing CU, Rohm M, Baysal-Temel E, Lehmann LH, Bauer R, Volz HC, Boutros M, Sohn D, Sticht C, Gretz N, Eichelbaum K, Werner T, Hirt MN, Eschenhagen T, Müller-Decker K, Strobel O, Hackert T, Krijgsveld J, Katus HA, Berriel Diaz M, Backs J, and Herzig S
- Published
- 2020
- Full Text
- View/download PDF
26. Hepatic Rab24 controls blood glucose homeostasis via improving mitochondrial plasticity.
- Author
-
Seitz S, Kwon Y, Hartleben G, Jülg J, Sekar R, Krahmer N, Najafi B, Loft A, Gancheva S, Stemmer K, Feuchtinger A, Hrabe de Angelis M, Müller TD, Mann M, Blüher M, Roden M, Berriel Diaz M, Behrends C, Gilleron J, Herzig S, and Zeigerer A
- Subjects
- Adiposity, Adult, Animals, Autophagy, Cholesterol blood, Female, Homeostasis, Humans, Lipid Metabolism genetics, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Non-alcoholic Fatty Liver Disease metabolism, Obesity metabolism, Up-Regulation, rab GTP-Binding Proteins genetics, Blood Glucose metabolism, Mitochondria, Liver metabolism, rab GTP-Binding Proteins metabolism
- Abstract
Non-alcoholic fatty liver disease (NAFLD) represents a key feature of obesity-related type 2 diabetes with increasing prevalence worldwide. To our knowledge, no treatment options are available to date, paving the way for more severe liver damage, including cirrhosis and hepatocellular carcinoma. Here, we show an unexpected function for an intracellular trafficking regulator, the small Rab GTPase Rab24, in mitochondrial fission and activation, which has an immediate impact on hepatic and systemic energy homeostasis. RAB24 is highly upregulated in the livers of obese patients with NAFLD and positively correlates with increased body fat in humans. Liver-selective inhibition of Rab24 increases autophagic flux and mitochondrial connectivity, leading to a strong improvement in hepatic steatosis and a reduction in serum glucose and cholesterol levels in obese mice. Our study highlights a potential therapeutic application of trafficking regulators, such as RAB24, for NAFLD and establishes a conceptual functional connection between intracellular transport and systemic metabolic dysfunction.
- Published
- 2019
- Full Text
- View/download PDF
27. Cancer Cachexia: More Than Skeletal Muscle Wasting.
- Author
-
Schmidt SF, Rohm M, Herzig S, and Berriel Diaz M
- Subjects
- Antineoplastic Agents pharmacology, Cachexia etiology, Cachexia mortality, Cachexia prevention & control, Combined Modality Therapy methods, Humans, Muscle, Skeletal physiopathology, Neoplasms drug therapy, Neoplasms mortality, Neoplasms physiopathology, Paraneoplastic Syndromes etiology, Paraneoplastic Syndromes mortality, Paraneoplastic Syndromes prevention & control, Quality of Life, Treatment Outcome, Antineoplastic Agents therapeutic use, Cachexia physiopathology, Neoplasms complications, Nutritional Support methods, Paraneoplastic Syndromes physiopathology
- Abstract
Cancer cachexia is a multifactorial condition characterized by body weight loss that negatively affects quality of life and survival of patients with cancer. Despite the clinical relevance, there is currently no defined standard of care to effectively counteract cancer-associated progressive tissue wasting. Skeletal muscle atrophy represents the main manifestation of cancer cachexia. However, cancer cachexia is increasingly seen as a systemic phenomenon affecting and/or influenced by various organs. Here, we describe recent developments elucidating the roles of different tissues as well as tissue crosstalk in this wasting syndrome, including potential links to other cancer-associated morbidities. A more comprehensive understanding of cancer cachexia etiology and heterogeneity may enable the development of intervention strategies to prevent or reverse this devastating condition., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
28. Acetyl-CoA Carboxylase 1-Dependent Protein Acetylation Controls Breast Cancer Metastasis and Recurrence.
- Author
-
Rios Garcia M, Steinbauer B, Srivastava K, Singhal M, Mattijssen F, Maida A, Christian S, Hess-Stumpp H, Augustin HG, Müller-Decker K, Nawroth PP, Herzig S, and Berriel Diaz M
- Subjects
- Acetyl-CoA Carboxylase genetics, Acetylation, Animals, Disease Models, Animal, Female, HEK293 Cells, Humans, Leptin metabolism, MCF-7 Cells, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Neoplasm Metastasis, Neoplasm Recurrence, Local metabolism, Tissue Array Analysis, Acetyl-CoA Carboxylase metabolism, Breast Neoplasms metabolism, Breast Neoplasms pathology, Lung Neoplasms secondary, Neoplasm Recurrence, Local pathology
- Abstract
Breast tumor recurrence and metastasis represent the main causes of cancer-related death in women, and treatments are still lacking. Here, we define the lipogenic enzyme acetyl-CoA carboxylase (ACC) 1 as a key player in breast cancer metastasis. ACC1 phosphorylation was increased in invading cells both in murine and human breast cancer, serving as a point of convergence for leptin and transforming growth factor (TGF) β signaling. ACC1 phosphorylation was mediated by TGFβ-activated kinase (TAK) 1, and ACC1 inhibition was indispensable for the elevation of cellular acetyl-CoA, the subsequent increase in Smad2 transcription factor acetylation and activation, and ultimately epithelial-mesenchymal transition and metastasis induction. ACC1 deficiency worsened tumor recurrence upon primary tumor resection in mice, and ACC1 phosphorylation levels correlated with metastatic potential in breast and lung cancer patients. Given the demonstrated effectiveness of anti-leptin receptor antibody treatment in halting ACC1-dependent tumor invasiveness, our work defines a "metabolocentric" approach in metastatic breast cancer therapy., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
29. A Hepatic GAbp-AMPK Axis Links Inflammatory Signaling to Systemic Vascular Damage.
- Author
-
Niopek K, Üstünel BE, Seitz S, Sakurai M, Zota A, Mattijssen F, Wang X, Sijmonsma T, Feuchter Y, Gail AM, Leuchs B, Niopek D, Staufer O, Brune M, Sticht C, Gretz N, Müller-Decker K, Hammes HP, Nawroth P, Fleming T, Conkright MD, Blüher M, Zeigerer A, Herzig S, and Berriel Diaz M
- Subjects
- AMP-Activated Protein Kinase Kinases, Animals, Atherosclerosis etiology, Atherosclerosis pathology, Cell Line, Cells, Cultured, Cholesterol metabolism, GA-Binding Protein Transcription Factor chemistry, Hypercholesterolemia complications, Male, Mice, Mice, Inbred C57BL, Protein Multimerization, Protein Subunits chemistry, Protein Subunits metabolism, Reactive Oxygen Species metabolism, Tumor Necrosis Factor-alpha metabolism, Atherosclerosis metabolism, GA-Binding Protein Transcription Factor metabolism, Hepatocytes metabolism, Hypercholesterolemia metabolism, Protein Kinases metabolism, Signal Transduction
- Abstract
Increased pro-inflammatory signaling is a hallmark of metabolic dysfunction in obesity and diabetes. Although both inflammatory and energy substrate handling processes represent critical layers of metabolic control, their molecular integration sites remain largely unknown. Here, we identify the heterodimerization interface between the α and β subunits of transcription factor GA-binding protein (GAbp) as a negative target of tumor necrosis factor alpha (TNF-α) signaling. TNF-α prevented GAbpα and β complex formation via reactive oxygen species (ROS), leading to the non-energy-dependent transcriptional inactivation of AMP-activated kinase (AMPK) β1, which was identified as a direct hepatic GAbp target. Impairment of AMPKβ1, in turn, elevated downstream cellular cholesterol biosynthesis, and hepatocyte-specific ablation of GAbpα induced systemic hypercholesterolemia and early macro-vascular lesion formation in mice. As GAbpα and AMPKβ1 levels were also found to correlate in obese human patients, the ROS-GAbp-AMPK pathway may represent a key component of a hepato-vascular axis in diabetic long-term complications., (Copyright © 2017 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
30. In vivo assessment of cold stimulation effects on the fat fraction of brown adipose tissue using DIXON MRI.
- Author
-
Stahl V, Maier F, Freitag MT, Floca RO, Berger MC, Umathum R, Berriel Diaz M, Herzig S, Weber MA, Dimitrakopoulou-Strauss A, Rink K, Bachert P, Ladd ME, and Nagel AM
- Subjects
- Adult, Algorithms, Body Temperature physiology, Female, Humans, Image Enhancement methods, Male, Organ Size, Reproducibility of Results, Sensitivity and Specificity, Adipose Tissue, Brown anatomy & histology, Adipose Tissue, Brown physiology, Adiposity physiology, Cold Temperature, Cold-Shock Response physiology, Image Interpretation, Computer-Assisted methods, Magnetic Resonance Imaging methods
- Abstract
Purpose: To evaluate the volume and changes of human brown adipose tissue (BAT) in vivo following exposure to cold using magnetic resonance imaging (MRI)., Materials and Methods: The clavicular region of 10 healthy volunteers was examined with a 3T MRI system. One volunteer participated twice. A cooling vest that was circulated with temperature-controlled water was used to expose each volunteer to a cold environment. Three different water temperature phases were employed: baseline (23°C, 20 min), cooling (12°C, 90 min), and a final warming phase (37°C, 30 min). Temperatures of the water in the circuit, of the body, and at the back skin of the volunteers were monitored with fiberoptic temperature probes. Applying the 2-point DIXON pulse sequence every 5 minutes, fat fraction (FF) maps were determined and evaluated over time to distinguish between brown and white adipose tissue., Results: Temperature measurements showed a decrease of 3.8 ± 1.0°C of the back skin temperature, while the body temperature stayed constant at 37.2 ± 0.9°C. Focusing on the two interscapular BAT depots, a mean FF decrease of -2.9 ± 2.0%/h (P < 0.001) was detected during cold stimulation in a mean absolute volume of 1.31 ± 1.43 ml. Also, a correlation of FF decrease to back skin temperature decrease was observed in all volunteers (correlation coefficients: |r| = [0.51; 0.99])., Conclusion: We found that FF decreases in BAT begin immediately with mild cooling of the body and continue during long-time cooling., Level of Evidence: 2 J. Magn. Reson. Imaging 2017;45:369-380., (© 2016 International Society for Magnetic Resonance in Medicine.)
- Published
- 2017
- Full Text
- View/download PDF
31. Control of diabetic hyperglycaemia and insulin resistance through TSC22D4.
- Author
-
Ekim Üstünel B, Friedrich K, Maida A, Wang X, Krones-Herzig A, Seibert O, Sommerfeld A, Jones A, Sijmonsma TP, Sticht C, Gretz N, Fleming T, Nawroth PP, Stremmel W, Rose AJ, Berriel-Diaz M, Blüher M, and Herzig S
- Subjects
- Animals, Cell Line, Diabetes Mellitus, Type 2 blood, Female, Gene Expression Regulation, Humans, Hyperglycemia blood, Lipocalins genetics, Lipocalins metabolism, Liver metabolism, Male, Mice, Inbred C57BL, Mice, Knockout, Transcription Factors metabolism, Blood Glucose metabolism, Diabetes Mellitus, Type 2 genetics, Hyperglycemia genetics, Insulin Resistance genetics, Transcription Factors genetics
- Abstract
Obesity-related insulin resistance represents the core component of the metabolic syndrome, promoting glucose intolerance, pancreatic beta cell failure and type 2 diabetes. Efficient and safe insulin sensitization and glucose control remain critical therapeutic aims to prevent diabetic late complications Here, we identify transforming growth factor beta-like stimulated clone (TSC) 22 D4 as a molecular determinant of insulin signalling and glucose handling. Hepatic TSC22D4 inhibition both prevents and reverses hyperglycaemia, glucose intolerance and insulin resistance in diabetes mouse models. TSC22D4 exerts its effects on systemic glucose homeostasis-at least in part-through the direct transcriptional regulation of the small secretory protein lipocalin 13 (LCN13). Human diabetic patients display elevated hepatic TSC22D4 expression, which correlates with decreased insulin sensitivity, hyperglycaemia and LCN13 serum levels. Our results establish TSC22D4 as a checkpoint in systemic glucose metabolism in both mice and humans, and propose TSC22D4 inhibition as an insulin sensitizing option in diabetes therapy.
- Published
- 2016
- Full Text
- View/download PDF
32. An AMP-activated protein kinase-stabilizing peptide ameliorates adipose tissue wasting in cancer cachexia in mice.
- Author
-
Rohm M, Schäfer M, Laurent V, Üstünel BE, Niopek K, Algire C, Hautzinger O, Sijmonsma TP, Zota A, Medrikova D, Pellegata NS, Ryden M, Kulyte A, Dahlman I, Arner P, Petrovic N, Cannon B, Amri EZ, Kemp BE, Steinberg GR, Janovska P, Kopecky J, Wolfrum C, Blüher M, Berriel Diaz M, and Herzig S
- Subjects
- AMP-Activated Protein Kinases pharmacology, Adipocytes, White metabolism, Adipose Tissue, White metabolism, Animals, Apoptosis Regulatory Proteins metabolism, Cachexia etiology, Cells, Cultured, In Vitro Techniques, Lipogenesis drug effects, Lipolysis drug effects, Mice, Neoplasms complications, Thermogenesis drug effects, Uncoupling Protein 1 drug effects, Uncoupling Protein 1 metabolism, AMP-Activated Protein Kinases metabolism, Adipocytes, White drug effects, Adipose Tissue, White drug effects, Apoptosis Regulatory Proteins drug effects, Cachexia metabolism, Lipid Metabolism drug effects, Neoplasms metabolism, Peptide Fragments pharmacology
- Abstract
Cachexia represents a fatal energy-wasting syndrome in a large number of patients with cancer that mostly results in a pathological loss of skeletal muscle and adipose tissue. Here we show that tumor cell exposure and tumor growth in mice triggered a futile energy-wasting cycle in cultured white adipocytes and white adipose tissue (WAT), respectively. Although uncoupling protein 1 (Ucp1)-dependent thermogenesis was dispensable for tumor-induced body wasting, WAT from cachectic mice and tumor-cell-supernatant-treated adipocytes were consistently characterized by the simultaneous induction of both lipolytic and lipogenic pathways. Paradoxically, this was accompanied by an inactivated AMP-activated protein kinase (Ampk), which is normally activated in peripheral tissues during states of low cellular energy. Ampk inactivation correlated with its degradation and with upregulation of the Ampk-interacting protein Cidea. Therefore, we developed an Ampk-stabilizing peptide, ACIP, which was able to ameliorate WAT wasting in vitro and in vivo by shielding the Cidea-targeted interaction surface on Ampk. Thus, our data establish the Ucp1-independent remodeling of adipocyte lipid homeostasis as a key event in tumor-induced WAT wasting, and we propose the ACIP-dependent preservation of Ampk integrity in the WAT as a concept in future therapies for cachexia.
- Published
- 2016
- Full Text
- View/download PDF
33. Fasting-induced liver GADD45β restrains hepatic fatty acid uptake and improves metabolic health.
- Author
-
Fuhrmeister J, Zota A, Sijmonsma TP, Seibert O, Cıngır Ş, Schmidt K, Vallon N, de Guia RM, Niopek K, Berriel Diaz M, Maida A, Blüher M, Okun JG, Herzig S, and Rose AJ
- Subjects
- Animals, Fatty Acid-Binding Proteins metabolism, Mice, Mice, Knockout, Cell Cycle Proteins metabolism, Fasting, Fatty Acids metabolism, Liver metabolism, Nuclear Proteins metabolism
- Abstract
Recent studies have demonstrated that repeated short-term nutrient withdrawal (i.e. fasting) has pleiotropic actions to promote organismal health and longevity. Despite this, the molecular physiological mechanisms by which fasting is protective against metabolic disease are largely unknown. Here, we show that, metabolic control, particularly systemic and liver lipid metabolism, is aberrantly regulated in the fasted state in mouse models of metabolic dysfunction. Liver transcript assays between lean/healthy and obese/diabetic mice in fasted and fed states uncovered "growth arrest and DNA damage-inducible" GADD45β as a dysregulated gene transcript during fasting in several models of metabolic dysfunction including ageing, obesity/pre-diabetes and type 2 diabetes, in both mice and humans. Using whole-body knockout mice as well as liver/hepatocyte-specific gain- and loss-of-function strategies, we revealed a role for liver GADD45β in the coordination of liver fatty acid uptake, through cytoplasmic retention of FABP1, ultimately impacting obesity-driven hyperglycaemia. In summary, fasting stress-induced GADD45β represents a liver-specific molecular event promoting adaptive metabolic function., (© 2016 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2016
- Full Text
- View/download PDF
34. Biological Mechanisms for the Effect of Obesity on Cancer Risk: Experimental Evidence.
- Author
-
Berriel Diaz M, Herzig S, and Schafmeier T
- Subjects
- Adipose Tissue physiopathology, Adiposity, Animals, Cell Transformation, Neoplastic pathology, Energy Metabolism, Gastrointestinal Microbiome, Humans, Inflammation Mediators metabolism, Neoplasms metabolism, Neoplasms pathology, Obesity metabolism, Obesity physiopathology, Risk Factors, Signal Transduction, Adipokines metabolism, Adipose Tissue metabolism, Biomarkers, Tumor metabolism, Cell Transformation, Neoplastic metabolism, Neoplasms etiology, Obesity complications
- Abstract
Multiple epidemiological studies demonstrated that overweight and obesity significantly increase the risk of several types of cancer. As the prevalence of obesity is dramatically rising, it is expected that it will represent one of the major lifestyle-associated risk factors for cancer development in the near future. Numerous recent studies expanded knowledge about key players and pathways, which are deregulated in the obese state and potentially promote cancer initiation, progression and aggressiveness via remote and local effects. These players include (but are not limited to) insulin/IGF, adipokines and inflammatory signaling molecules as well as metabolites. Nevertheless, the detailed mechanisms linking obesity and malignant transformation at the systemic, cellular and molecular level still demand further investigation. Additionally, dysfunctional molecular metabolic pathways appear to be specific for distinct cancer entities, thereby yet precluding definition of a common principle. This chapter will present an overview of the current knowledge of molecular nodes linking obesity and cancer and will briefly touch upon potential therapy options addressing metabolic cancer etiologies.
- Published
- 2016
- Full Text
- View/download PDF
35. Ataxin-10 is part of a cachexokine cocktail triggering cardiac metabolic dysfunction in cancer cachexia.
- Author
-
Schäfer M, Oeing CU, Rohm M, Baysal-Temel E, Lehmann LH, Bauer R, Volz HC, Boutros M, Sohn D, Sticht C, Gretz N, Eichelbaum K, Werner T, Hirt MN, Eschenhagen T, Müller-Decker K, Strobel O, Hackert T, Krijgsveld J, Katus HA, Berriel Diaz M, Backs J, and Herzig S
- Abstract
Objectives: Cancer cachexia affects the majority of tumor patients and significantly contributes to high mortality rates in these subjects. Despite its clinical importance, the identity of tumor-borne signals and their impact on specific peripheral organ systems, particularly the heart, remain mostly unknown., Methods and Results: By combining differential colon cancer cell secretome profiling with large-scale cardiomyocyte phenotyping, we identified a signature panel of seven "cachexokines", including Bridging integrator 1, Syntaxin 7, Multiple inositol-polyphosphate phosphatase 1, Glucosidase alpha acid, Chemokine ligand 2, Adamts like 4, and Ataxin-10, which were both sufficient and necessary to trigger cardiac atrophy and aberrant fatty acid metabolism in cardiomyocytes. As a prototypical example, engineered secretion of Ataxin-10 from non-cachexia-inducing cells was sufficient to induce cachexia phenotypes in cardiomyocytes, correlating with elevated Ataxin-10 serum levels in murine and human cancer cachexia models., Conclusions: As Ataxin-10 serum levels were also found to be elevated in human cachectic cancer patients, the identification of Ataxin-10 as part of a cachexokine cocktail now provides a rational approach towards personalized predictive, diagnostic and therapeutic measures in cancer cachexia.
- Published
- 2015
- Full Text
- View/download PDF
36. PPP2R5C Couples Hepatic Glucose and Lipid Homeostasis.
- Author
-
Cheng YS, Seibert O, Klöting N, Dietrich A, Straßburger K, Fernández-Veledo S, Vendrell JJ, Zorzano A, Blüher M, Herzig S, Berriel Diaz M, and Teleman AA
- Subjects
- AMP-Activated Protein Kinases genetics, Animals, Dietary Carbohydrates metabolism, Glucose metabolism, Hepatocytes metabolism, Humans, Insulin Resistance genetics, Lipogenesis genetics, Liver metabolism, Mice, Obesity pathology, Sterol Regulatory Element Binding Protein 1 genetics, Energy Metabolism genetics, Lipid Metabolism genetics, Obesity genetics, Protein Phosphatase 2 genetics
- Abstract
In mammals, the liver plays a central role in maintaining carbohydrate and lipid homeostasis by acting both as a major source and a major sink of glucose and lipids. In particular, when dietary carbohydrates are in excess, the liver converts them to lipids via de novo lipogenesis. The molecular checkpoints regulating the balance between carbohydrate and lipid homeostasis, however, are not fully understood. Here we identify PPP2R5C, a regulatory subunit of PP2A, as a novel modulator of liver metabolism in postprandial physiology. Inactivation of PPP2R5C in isolated hepatocytes leads to increased glucose uptake and increased de novo lipogenesis. These phenotypes are reiterated in vivo, where hepatocyte specific PPP2R5C knockdown yields mice with improved systemic glucose tolerance and insulin sensitivity, but elevated circulating triglyceride levels. We show that modulation of PPP2R5C levels leads to alterations in AMPK and SREBP-1 activity. We find that hepatic levels of PPP2R5C are elevated in human diabetic patients, and correlate with obesity and insulin resistance in these subjects. In sum, our data suggest that hepatic PPP2R5C represents an important factor in the functional wiring of energy metabolism and the maintenance of a metabolically healthy state.
- Published
- 2015
- Full Text
- View/download PDF
37. Transcriptional co-factor Transducin beta-like (TBL) 1 acts as a checkpoint in pancreatic cancer malignancy.
- Author
-
Stoy C, Sundaram A, Rios Garcia M, Wang X, Seibert O, Zota A, Wendler S, Männle D, Hinz U, Sticht C, Muciek M, Gretz N, Rose AJ, Greiner V, Hofmann TG, Bauer A, Hoheisel J, Berriel Diaz M, Gaida MM, Werner J, Schafmeier T, Strobel O, and Herzig S
- Subjects
- Animals, Gene Expression Profiling, Humans, Mice, Survival Analysis, Transducin deficiency, Carcinoma, Pancreatic Ductal pathology, Pancreatic Neoplasms pathology, Transducin metabolism
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer fatalities in Western societies, characterized by high metastatic potential and resistance to chemotherapy. Critical molecular mechanisms of these phenotypical features still remain unknown, thus hampering the development of effective prognostic and therapeutic measures in PDAC. Here, we show that transcriptional co-factor Transducin beta-like (TBL) 1 was over-expressed in both human and murine PDAC. Inactivation of TBL1 in human and mouse pancreatic cancer cells reduced cellular proliferation and invasiveness, correlating with diminished glucose uptake, glycolytic flux, and oncogenic PI3 kinase signaling which in turn could rescue TBL1 deficiency-dependent phenotypes. TBL1 deficiency both prevented and reversed pancreatic tumor growth, mediated transcriptional PI3 kinase inhibition, and increased chemosensitivity of PDAC cells in vivo. As TBL1 mRNA levels were also found to correlate with PI3 kinase levels and overall survival in a cohort of human PDAC patients, TBL1 was identified as a checkpoint in the malignant behavior of pancreatic cancer and its expression may serve as a novel molecular target in the treatment of human PDAC., (© 2015 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2015
- Full Text
- View/download PDF
38. microRNA-379 couples glucocorticoid hormones to dysfunctional lipid homeostasis.
- Author
-
de Guia RM, Rose AJ, Sommerfeld A, Seibert O, Strzoda D, Zota A, Feuchter Y, Krones-Herzig A, Sijmonsma T, Kirilov M, Sticht C, Gretz N, Dallinga-Thie G, Diederichs S, Klöting N, Blüher M, Berriel Diaz M, and Herzig S
- Subjects
- Animals, Cell Line, Female, Gene Silencing, Glucocorticoids genetics, Humans, Lipoproteins, VLDL genetics, Lipoproteins, VLDL metabolism, Liver pathology, Male, Mice, Mice, Obese, MicroRNAs genetics, Obesity genetics, Triglycerides genetics, Triglycerides metabolism, Glucocorticoids metabolism, Lipid Metabolism, Liver metabolism, MicroRNAs metabolism, Obesity metabolism
- Abstract
In mammals, glucocorticoids (GCs) and their intracellular receptor, the glucocorticoid receptor (GR), represent critical checkpoints in the endocrine control of energy homeostasis. Indeed, aberrant GC action is linked to severe metabolic stress conditions as seen in Cushing's syndrome, GC therapy and certain components of the Metabolic Syndrome, including obesity and insulin resistance. Here, we identify the hepatic induction of the mammalian conserved microRNA (miR)-379/410 genomic cluster as a key component of GC/GR-driven metabolic dysfunction. Particularly, miR-379 was up-regulated in mouse models of hyperglucocorticoidemia and obesity as well as human liver in a GC/GR-dependent manner. Hepatocyte-specific silencing of miR-379 substantially reduced circulating very-low-density lipoprotein (VLDL)-associated triglyceride (TG) levels in healthy mice and normalized aberrant lipid profiles in metabolically challenged animals, mediated through miR-379 effects on key receptors in hepatic TG re-uptake. As hepatic miR-379 levels were also correlated with GC and TG levels in human obese patients, the identification of a GC/GR-controlled miRNA cluster not only defines a novel layer of hormone-dependent metabolic control but also paves the way to alternative miRNA-based therapeutic approaches in metabolic dysfunction., (© 2014 The Authors.)
- Published
- 2015
- Full Text
- View/download PDF
39. Transcriptional cofactor TBLR1 controls lipid mobilization in white adipose tissue.
- Author
-
Rohm M, Sommerfeld A, Strzoda D, Jones A, Sijmonsma TP, Rudofsky G, Wolfrum C, Sticht C, Gretz N, Zeyda M, Leitner L, Nawroth PP, Stulnig TM, Berriel Diaz M, Vegiopoulos A, and Herzig S
- Subjects
- 3T3-L1 Cells, Animals, Cyclic AMP metabolism, Diet, High-Fat, Fatty Acids, Nonesterified blood, Humans, Insulin Resistance, Lipolysis, Mice, Mice, Inbred C57BL, Mice, Knockout, Mice, Obese, Obesity metabolism, Obesity pathology, RNA Interference, RNA, Messenger metabolism, RNA, Small Interfering metabolism, Receptors, Adrenergic genetics, Receptors, Adrenergic metabolism, Receptors, Cytoplasmic and Nuclear antagonists & inhibitors, Receptors, Cytoplasmic and Nuclear genetics, Signal Transduction, Adipose Tissue, White metabolism, Lipid Mobilization physiology, Receptors, Cytoplasmic and Nuclear metabolism
- Abstract
Lipid mobilization (lipolysis) in white adipose tissue (WAT) critically controls lipid turnover and adiposity in humans. While the acute regulation of lipolysis has been studied in detail, the transcriptional determinants of WAT lipolytic activity remain still largely unexplored. Here we show that the genetic inactivation of transcriptional cofactor transducin beta-like-related 1(TBLR1) blunts the lipolytic response of white adipocytes through the impairment of cAMP-dependent signal transduction. Indeed, mice lacking TBLR1 in adipocytes are defective in fasting-induced lipid mobilization and, when placed on a high-fat-diet, show aggravated adiposity, glucose intolerance, and insulin resistance. TBLR1 levels are found to increase under lipolytic conditions in WAT of both human patients and mice, correlating with serum free fatty acids (FFAs). As a critical regulator of WAT cAMP signaling and lipid mobilization, proper activity of TBLR1 in adipocytes might thus represent a critical molecular checkpoint for the prevention of metabolic dysfunction in subjects with obesity-related disorders., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
40. Detecting endogenous SUMO targets in mammalian cells and tissues.
- Author
-
Becker J, Barysch SV, Karaca S, Dittner C, Hsiao HH, Berriel Diaz M, Herzig S, Urlaub H, and Melchior F
- Subjects
- Amino Acid Sequence, Animals, Antibodies, Monoclonal immunology, Cells, Cultured, Electrophoresis, Polyacrylamide Gel, Humans, Mammals, Molecular Sequence Data, Sequence Homology, Amino Acid, Small Ubiquitin-Related Modifier Proteins chemistry, Small Ubiquitin-Related Modifier Proteins immunology, Small Ubiquitin-Related Modifier Proteins metabolism
- Abstract
SUMOylation is an essential modification that regulates hundreds of proteins in eukaryotic cells. Owing to its dynamic nature and low steady-state levels, endogenous SUMOylation is challenging to detect. Here, we present a method that allows efficient enrichment and identification of endogenous targets of SUMO1 and the nearly identical SUMO2 and 3 (SUMO 2/3) from vertebrate cells and complex organ tissue. Using monoclonal antibodies for which we mapped the epitope, we enriched SUMOylated proteins by immunoprecipitation and peptide elution. We used this approach in combination with MS to identify SUMOylated proteins, which resulted in the first direct comparison of the endogenous SUMO1- and SUMO2/3-modified proteome in mammalian cells, to our knowledge. This protocol provides an affordable and feasible tool to investigate endogenous SUMOylation in primary cells, tissues and organs, and it will facilitate understanding of SUMO's role in physiology and disease.
- Published
- 2013
- Full Text
- View/download PDF
41. Selective enrichment of newly synthesized proteins for quantitative secretome analysis.
- Author
-
Eichelbaum K, Winter M, Berriel Diaz M, Herzig S, and Krijgsveld J
- Subjects
- Cell Line, Culture Media, Culture Media, Serum-Free, Hepatocytes drug effects, Hepatocytes metabolism, Humans, Kinetics, Lipopolysaccharides pharmacology, Macrophage Activation drug effects, Protein Interaction Maps drug effects, Click Chemistry methods, Isotope Labeling methods, Protein Biosynthesis drug effects, Proteins metabolism
- Abstract
Secreted proteins constitute a large and biologically important subset of proteins that are involved in cellular communication, adhesion and migration. Yet secretomes are understudied because of technical limitations in the detection of low-abundance proteins against a background of serum-containing media. Here we introduce a method that combines click chemistry and pulsed stable isotope labeling with amino acids in cell culture to selectively enrich and quantify secreted proteins. The combination of these two labeling approaches allows cells to be studied irrespective of the complexity of the background proteins. We provide an in-depth and differential secretome analysis of various cell lines and primary cells, quantifying secreted factors, including cytokines, chemokines and growth factors. In addition, we reveal that serum starvation has a marked effect on secretome composition. We also analyze the kinetics of protein secretion by macrophages in response to lipopolysaccharides.
- Published
- 2012
- Full Text
- View/download PDF
42. Functional inactivation of the genome-wide association study obesity gene neuronal growth regulator 1 in mice causes a body mass phenotype.
- Author
-
Lee AW, Hengstler H, Schwald K, Berriel-Diaz M, Loreth D, Kirsch M, Kretz O, Haas CA, de Angelis MH, Herzig S, Brümmendorf T, Klingenspor M, Rathjen FG, Rozman J, Nicholson G, Cox RD, and Schäfer MK
- Subjects
- Alleles, Animals, Body Height genetics, Cell Adhesion, Cell Line, Diet, High-Fat adverse effects, Eating genetics, Endoplasmic Reticulum metabolism, Energy Metabolism genetics, Female, Gene Knockout Techniques, Genotype, Humans, Hypothalamus cytology, Hypothalamus metabolism, Male, Membrane Proteins metabolism, Mice, Motor Activity genetics, Nerve Tissue Proteins metabolism, Neurites metabolism, Obesity metabolism, Obesity pathology, Obesity physiopathology, Phenotype, Body Weight genetics, Gene Silencing, Genome-Wide Association Study, Membrane Proteins deficiency, Membrane Proteins genetics, Nerve Tissue Proteins deficiency, Nerve Tissue Proteins genetics, Obesity genetics
- Abstract
To date, genome-wide association studies (GWAS) have identified at least 32 novel loci for obesity and body mass-related traits. However, the causal genetic variant and molecular mechanisms of specific susceptibility genes in relation to obesity are yet to be fully confirmed and characterised. Here, we examined whether the candidate gene NEGR1 encoding the neuronal growth regulator 1, also termed neurotractin or Kilon, accounts for the obesity association. To characterise the function of NEGR1 for body weight control in vivo, we generated two novel mutant mouse lines, including a constitutive NEGR1-deficient mouse line as well as an ENU-mutagenised line carrying a loss-of-function mutation (Negr1-I87N) and performed metabolic phenotypic analyses. Ablation of NEGR1 results in a small but steady reduction of body mass in both mutant lines, accompanied with a small reduction in body length in the Negr1-I87N mutants. Magnetic resonance scanning reveals that the reduction of body mass in Negr1-I87N mice is due to a reduced proportion of lean mass. Negr1-I87N mutants display reduced food intake and physical activity while normalised energy expenditure remains unchanged. Expression analyses confirmed the brain-specific distribution of NEGR1 including strong expression in the hypothalamus. In vitro assays show that NEGR1 promotes cell-cell adhesion and neurite growth of hypothalamic neurons. Our results indicate a role of NEGR1 in the control of body weight and food intake. This study provides evidence that supports the link of the GWAS candidate gene NEGR1 with body weight control.
- Published
- 2012
- Full Text
- View/download PDF
43. Control of adipose tissue inflammation through TRB1.
- Author
-
Ostertag A, Jones A, Rose AJ, Liebert M, Kleinsorg S, Reimann A, Vegiopoulos A, Berriel Diaz M, Strzoda D, Yamamoto M, Satoh T, Akira S, and Herzig S
- Subjects
- Adipocytes physiology, Animals, Cell Line, Chromatin physiology, DNA Primers, Humans, Kidney embryology, Male, Mice, Mice, Inbred C57BL, Protein Serine-Threonine Kinases metabolism, RNA genetics, RNA, Messenger genetics, Reverse Transcriptase Polymerase Chain Reaction, Transfection, Adipose Tissue physiopathology, Inflammation physiopathology, Protein Serine-Threonine Kinases genetics
- Abstract
Objective: Based on its role as an energy storage compartment and endocrine organ, white adipose tissue (WAT) fulfills a critical function in the maintenance of whole-body energy homeostasis. Indeed, WAT dysfunction is connected to obesity-related type 2 diabetes triggered at least partly by an inflammatory response in adipocytes. The pseudokinase tribbles (TRB) 3 has been identified by us and others as a critical regulator of hepatic glucose homeostasis in type 2 diabetes and WAT lipid homeostasis. Therefore, this study aimed to test the hypothesis that the TRB gene family fulfills broader functions in the integration of metabolic and inflammatory pathways in various tissues., Research Design and Methods: To determine the role of TRB family members for WAT function, we profiled the expression patterns of TRB13 under healthy and metabolic stress conditions. The differentially expressed TRB1 was functionally characterized in loss-of-function animal and primary adipocyte models., Results: Here, we show that the expression of TRB1 was specifically upregulated during acute and chronic inflammation in WAT of mice. Deficiency of TRB1 was found to impair cytokine gene expression in white adipocytes and to protect against high-fat diet-induced obesity. In adipocytes, TRB1 served as a nuclear transcriptional coactivator for the nuclear factor kappaB subunit RelA, thereby promoting the induction of proinflammatory cytokines in these cells., Conclusions: As inflammation is typically seen in sepsis, insulin resistance, and obesity-related type 2 diabetes, the dual role of TRB1 as both a target and a (co) activator of inflammatory signaling might provide a molecular rationale for the amplification of proinflammatory responses in WAT in these subjects.
- Published
- 2010
- Full Text
- View/download PDF
44. Cyclooxygenase-2 controls energy homeostasis in mice by de novo recruitment of brown adipocytes.
- Author
-
Vegiopoulos A, Müller-Decker K, Strzoda D, Schmitt I, Chichelnitskiy E, Ostertag A, Berriel Diaz M, Rozman J, Hrabe de Angelis M, Nüsing RM, Meyer CW, Wahli W, Klingenspor M, and Herzig S
- Subjects
- Adipocytes, Brown cytology, Adipogenesis, Adipose Tissue, Adipose Tissue, Brown cytology, Adipose Tissue, White enzymology, Adrenergic beta-3 Receptor Agonists, Adrenergic beta-Agonists pharmacology, Animals, Body Weight, Dietary Fats administration & dosage, Dioxoles pharmacology, Female, Gene Expression Regulation, Enzymologic, Homeostasis, Male, Mesenchymal Stem Cells cytology, Mice, Mice, Inbred C57BL, Mice, Obese, Mice, Transgenic, Norepinephrine metabolism, Obesity etiology, Obesity prevention & control, Oxygen Consumption, Receptors, Adrenergic, beta-3 metabolism, Signal Transduction, Adipocytes, Brown physiology, Adipose Tissue, Brown physiology, Adipose Tissue, White physiology, Cyclooxygenase 2 genetics, Cyclooxygenase 2 metabolism, Energy Metabolism, Prostaglandins metabolism, Thermogenesis
- Abstract
Obesity results from chronic energy surplus and excess lipid storage in white adipose tissue (WAT). In contrast, brown adipose tissue (BAT) efficiently burns lipids through adaptive thermogenesis. Studying mouse models, we show that cyclooxygenase (COX)-2, a rate-limiting enzyme in prostaglandin (PG) synthesis, is a downstream effector of beta-adrenergic signaling in WAT and is required for the induction of BAT in WAT depots. PG shifted the differentiation of defined mesenchymal progenitors toward a brown adipocyte phenotype. Overexpression of COX-2 in WAT induced de novo BAT recruitment in WAT, increased systemic energy expenditure, and protected mice against high-fat diet-induced obesity. Thus, COX-2 appears integral to de novo BAT recruitment, which suggests that the PG pathway regulates systemic energy homeostasis.
- Published
- 2010
- Full Text
- View/download PDF
45. Protein kinase G controls brown fat cell differentiation and mitochondrial biogenesis.
- Author
-
Haas B, Mayer P, Jennissen K, Scholz D, Berriel Diaz M, Bloch W, Herzig S, Fässler R, and Pfeifer A
- Subjects
- Adipogenesis, Animals, Cyclic GMP, Cyclic GMP-Dependent Protein Kinases deficiency, Insulin metabolism, Ion Channels, Mice, Mice, Knockout, Mitochondrial Proteins, Nitric Oxide, Uncoupling Protein 1, rho-Associated Kinases, rhoA GTP-Binding Protein, Adipose Tissue, Brown cytology, Cell Differentiation, Cyclic GMP-Dependent Protein Kinases physiology, Mitochondria metabolism
- Abstract
Brown adipose tissue (BAT) is a primary site of energy expenditure through thermogenesis, which is mediated by the uncoupling protein-1 (UCP-1) in mitochondria. Here, we show that protein kinase G (PKG) is essential for brown fat cell differentiation. Induction of adipogenic markers and fat storage was impaired in the absence of PKGI. Furthermore, PKGI mediated the ability of nitric oxide (NO) and guanosine 3',5'-monophosphate (cGMP) to induce mitochondrial biogenesis and increase the abundance of UCP-1. Mechanistically, we found that PKGI controlled insulin signaling in BAT by inhibiting the activity of RhoA and Rho-associated kinase (ROCK), thereby relieving the inhibitory effects of ROCK on insulin receptor substrate-1 and activating the downstream phosphoinositide 3-kinase-Akt cascade. Thus, PKGI links NO and cGMP signaling with the RhoA-ROCK and the insulin pathways, thereby controlling induction of adipogenic and thermogenic programs during brown fat cell differentiation.
- Published
- 2009
- Full Text
- View/download PDF
46. In vivo phosphoenolpyruvate carboxykinase promoter mapping identifies disrupted hormonal synergism as a target of inflammation during sepsis in mice.
- Author
-
Chichelnitskiy E, Vegiopoulos A, Berriel Diaz M, Ziegler A, Kleiman A, Rauch A, Tuckermann J, and Herzig S
- Subjects
- Animals, Cells, Cultured, Cyclic AMP physiology, Glucocorticoids physiology, Glucose metabolism, Liver metabolism, Male, Mice, Mice, Inbred C57BL, Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha, RNA, Messenger analysis, Response Elements, Signal Transduction, Trans-Activators physiology, Transcription Factors, Inflammation etiology, Phosphoenolpyruvate Carboxykinase (GTP) genetics, Promoter Regions, Genetic, Sepsis metabolism
- Abstract
Unlabelled: In mammals, proper maintenance of blood glucose levels within narrow limits is one of the most critical prerequisites for healthy energy homeostasis and body function. Consequently, hyper- and hypoglycemia represent hallmarks of severe metabolic pathologies, including type II diabetes and acute sepsis, respectively. Although the liver plays a crucial role in the control of systemic glucose homeostasis, the molecular mechanisms of aberrant hepatic glucose regulation under metabolic stress conditions remain largely unknown. Here we report the development of a liver-specific adenoviral in vivo system for monitoring promoter activity of the key gluconeogenic enzyme gene phosphoenolpyruvate carboxykinase (PEPCK) in mice. By employing in vivo promoter deletion technology, the glucocorticoid response unit (GRU) and the cyclic adenosine monophosphate (cAMP)-responsive element (CRE) were identified as critical cis-regulatory targets of proinflammatory signaling under septic conditions. In particular, both elements were found to be required for inhibition of PEPCK transcription during sepsis, thereby mediating endotoxic hypoglycemia. Indeed, expression of nuclear receptor cofactor peroxisome proliferator-activator receptor coactivator 1alpha (PGC-1alpha), the molecular mediator of GRU/CRE synergism on the PEPCK promoter, was found to be specifically repressed in septic liver, and restoration of PGC-1alpha in cytokine-exposed hepatocytes blunted the inhibitory effect of proinflammatory signaling on PEPCK gene expression., Conclusion: The dysregulation of hormonal synergism through the repression of PGC-1alpha as identified by in vivo promoter monitoring may provide a molecular rationale for hypoglycemia during sepsis, thereby highlighting the importance of hepatic glucose homeostasis for metabolic dysfunction in these patients.
- Published
- 2009
- Full Text
- View/download PDF
47. Positional cloning of zinc finger domain transcription factor Zfp69, a candidate gene for obesity-associated diabetes contributed by mouse locus Nidd/SJL.
- Author
-
Scherneck S, Nestler M, Vogel H, Blüher M, Block MD, Berriel Diaz M, Herzig S, Schulz N, Teichert M, Tischer S, Al-Hasani H, Kluge R, Schürmann A, and Joost HG
- Subjects
- Adipose Tissue metabolism, Animals, Chromosome Mapping, Diabetes Mellitus, Type 2 metabolism, Female, Humans, Male, Mice, Mice, Inbred C57BL, Mice, Obese, Mice, Transgenic, Obesity metabolism, Transcription Factors chemistry, Transcription Factors metabolism, Zinc Fingers, Cloning, Molecular, Diabetes Mellitus, Type 2 genetics, Obesity genetics, Quantitative Trait Loci, Transcription Factors genetics
- Abstract
Polygenic type 2 diabetes in mouse models is associated with obesity and results from a combination of adipogenic and diabetogenic alleles. Here we report the identification of a candidate gene for the diabetogenic effect of a QTL (Nidd/SJL, Nidd1) contributed by the SJL, NON, and NZB strains in outcross populations with New Zealand Obese (NZO) mice. A critical interval of distal chromosome 4 (2.1 Mbp) conferring the diabetic phenotype was identified by interval-specific congenic introgression of SJL into diabetes-resistant C57BL/6J, and subsequent reporter cross with NZO. Analysis of the 10 genes in the critical interval by sequencing, qRT-PCR, and RACE-PCR revealed a striking allelic variance of Zfp69 encoding zinc finger domain transcription factor 69. In NZO and C57BL/6J, a retrotransposon (IAPLTR1a) in intron 3 disrupted the gene by formation of a truncated mRNA that lacked the coding sequence for the KRAB (Krüppel-associated box) and Znf-C2H2 domains of Zfp69, whereas the diabetogenic SJL, NON, and NZB alleles generated a normal mRNA. When combined with the B6.V-Lep(ob) background, the diabetogenic Zfp69(SJL) allele produced hyperglycaemia, reduced gonadal fat, and increased plasma and liver triglycerides. mRNA levels of the human orthologue of Zfp69, ZNF642, were significantly increased in adipose tissue from patients with type 2 diabetes. We conclude that Zfp69 is the most likely candidate for the diabetogenic effect of Nidd/SJL, and that retrotransposon IAPLTR1a contributes substantially to the genetic heterogeneity of mouse strains. Expression of the transcription factor in adipose tissue may play a role in the pathogenesis of type 2 diabetes., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2009
- Full Text
- View/download PDF
48. Liver-specific loss of lipolysis-stimulated lipoprotein receptor triggers systemic hyperlipidemia in mice.
- Author
-
Narvekar P, Berriel Diaz M, Krones-Herzig A, Hardeland U, Strzoda D, Stöhr S, Frohme M, and Herzig S
- Subjects
- Animals, Apolipoproteins E deficiency, Blood Glucose metabolism, Cholesterol blood, Disease Models, Animal, Fatty Acids, Nonesterified blood, Ketone Bodies blood, Lipolysis, Lipoproteins, VLDL metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Mice, Obese, RNA Interference, Receptors, LDL deficiency, Receptors, LDL physiology, Reverse Transcriptase Polymerase Chain Reaction, Triglycerides blood, Diabetes Mellitus, Experimental physiopathology, Diabetes Mellitus, Type 2 physiopathology, Hyperlipidemias physiopathology, Liver physiology, Receptors, LDL genetics, Receptors, Lipoprotein physiology
- Abstract
Objective: In mammals, proper storage and distribution of lipids in and between tissues is essential for the maintenance of energy homeostasis. In contrast, aberrantly high levels of triglycerides in the blood ("hypertriglyceridemia") represent a hallmark of the metabolic syndrome and type 2 diabetes. As hypertriglyceridemia has been identified as an important risk factor for cardiovascular complications, in this study we aimed to identify molecular mechanisms in aberrant triglyceride elevation under these conditions., Research Design and Methods: To determine the importance of hepatic lipid handling for systemic dyslipidemia, we profiled the expression patterns of various hepatic lipid transporters and receptors under healthy and type 2 diabetic conditions. A differentially expressed lipoprotein receptor was functionally characterized by generating acute, liver-specific loss- and gain-of-function animal models., Results: We show that the hepatic expression of lipid transporter lipolysis-stimulated lipoprotein receptor (LSR) is specifically impaired in mouse models of obesity and type 2 diabetes and can be restored by leptin replacement. Experimental imitation of this pathophysiological situation by liver-specific knockdown of LSR promotes hypertriglyceridemia and elevated apolipoprotein (Apo)B and E serum levels in lean wild-type and ApoE knockout mice. In contrast, genetic restoration of LSR expression in obese animals to wild-type levels improves serum triglyceride levels and serum profiles in these mice., Conclusions: The dysregulation of hepatic LSR under obese and diabetic conditions may provide a molecular rationale for systemic dyslipidemia in type 2 diabetes and the metabolic syndrome and represent a novel target for alternative treatment strategies in these patients.
- Published
- 2009
- Full Text
- View/download PDF
49. The glucocorticoid receptor controls hepatic dyslipidemia through Hes1.
- Author
-
Lemke U, Krones-Herzig A, Berriel Diaz M, Narvekar P, Ziegler A, Vegiopoulos A, Cato AC, Bohl S, Klingmüller U, Screaton RA, Müller-Decker K, Kersten S, and Herzig S
- Subjects
- Animals, Basic Helix-Loop-Helix Transcription Factors genetics, Cells, Cultured, Disease Models, Animal, Histone Deacetylases metabolism, Homeodomain Proteins genetics, Male, Mice, Mice, Inbred C57BL, Mice, Obese, Obesity metabolism, Phenotype, Polymerase Chain Reaction methods, Promoter Regions, Genetic genetics, RNA, Messenger genetics, Receptors, Glucocorticoid deficiency, Transcription Factor HES-1, Transfection, Triglycerides metabolism, Basic Helix-Loop-Helix Transcription Factors metabolism, Fatty Liver metabolism, Homeodomain Proteins metabolism, Receptors, Glucocorticoid metabolism
- Abstract
Aberrant accumulation of lipids in the liver ("fatty liver" or hepatic steatosis) represents a hallmark of the metabolic syndrome and is tightly associated with obesity, type II diabetes, starvation, or glucocorticoid (GC) therapy. While fatty liver has been connected with numerous abnormalities of liver function, the molecular mechanisms of fatty liver development remain largely enigmatic. Here we show that liver-specific disruption of glucocorticoid receptor (GR) action improves the steatotic phenotype in fatty liver mouse models and leads to the induction of transcriptional repressor hairy enhancer of split 1 (Hes1) gene expression. The GR directly interferes with Hes1 promoter activity, triggering the recruitment of histone deacetylase (HDAC) activities to the Hes1 gene. Genetic restoration of hepatic Hes1 levels in steatotic animals normalizes hepatic triglyceride (TG) levels. As glucocorticoid action is increased during starvation, myotonic dystrophy, and Cushing's syndrome, the inhibition of Hes1 through the GR might explain the fatty liver phenotype in these subjects.
- Published
- 2008
- Full Text
- View/download PDF
50. Nuclear receptor cofactor receptor interacting protein 140 controls hepatic triglyceride metabolism during wasting in mice.
- Author
-
Berriel Diaz M, Krones-Herzig A, Metzger D, Ziegler A, Vegiopoulos A, Klingenspor M, Müller-Decker K, and Herzig S
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Animals, Cachexia physiopathology, Carcinoma, Hepatocellular metabolism, Carcinoma, Hepatocellular physiopathology, Cell Line, Cells, Cultured, Disease Models, Animal, Energy Metabolism physiology, Gene Expression Regulation, Homeostasis physiology, Humans, Liver microbiology, Liver physiopathology, Liver Neoplasms metabolism, Liver Neoplasms physiopathology, Male, Mice, Mice, Inbred C57BL, Nuclear Proteins genetics, Nuclear Receptor Interacting Protein 1, RNA Interference, Sepsis metabolism, Sepsis physiopathology, Transfection, Adaptor Proteins, Signal Transducing metabolism, Cachexia metabolism, Lipid Metabolism physiology, Liver metabolism, Nuclear Proteins metabolism, Triglycerides metabolism
- Abstract
Unlabelled: In mammals, triglycerides (TG) represent the most concentrated form of energy. Aberrant TG storage and availability are intimately linked to the negative energy balance under severe clinical conditions, such as starvation, sepsis, or cancer cachexia. Despite its crucial role for energy homeostasis, molecular key determinants of TG metabolism remain enigmatic. Here we show that the expression of nuclear receptor cofactor receptor interacting protein (RIP) 140 was induced in livers of starved, septic, and tumor-bearing mice. Liver-specific knockdown of RIP140 led to increased hepatic TG release and alleviated hepatic steatosis in tumor-bearing, cachectic animals. Indeed, hepatic RIP140 was found to control the expression of lipid-metabolizing genes in liver., Conclusion: By preventing the mobilization of hepatic TG stores, the induction of RIP140 in liver provides a molecular rationale for hepatic steatosis in starvation, sepsis, or cancer cachexia. Inhibition of hepatic RIP140 transcriptional activity might, thereby, provide an attractive adjunct scheme in the treatment of these conditions.
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.