Go to JCI Insight
  • About
  • Editors
  • Consulting Editors
  • For authors
  • Publication ethics
  • Publication alerts by email
  • Advertising
  • Job board
  • Contact
  • Clinical Research and Public Health
  • Current issue
  • Past issues
  • By specialty
    • COVID-19
    • Cardiology
    • Gastroenterology
    • Immunology
    • Metabolism
    • Nephrology
    • Neuroscience
    • Oncology
    • Pulmonology
    • Vascular biology
    • All ...
  • Videos
    • Conversations with Giants in Medicine
    • Video Abstracts
  • Reviews
    • View all reviews ...
    • Complement Biology and Therapeutics (May 2025)
    • Evolving insights into MASLD and MASH pathogenesis and treatment (Apr 2025)
    • Microbiome in Health and Disease (Feb 2025)
    • Substance Use Disorders (Oct 2024)
    • Clonal Hematopoiesis (Oct 2024)
    • Sex Differences in Medicine (Sep 2024)
    • Vascular Malformations (Apr 2024)
    • View all review series ...
  • Viewpoint
  • Collections
    • In-Press Preview
    • Clinical Research and Public Health
    • Research Letters
    • Letters to the Editor
    • Editorials
    • Commentaries
    • Editor's notes
    • Reviews
    • Viewpoints
    • 100th anniversary
    • Top read articles

  • Current issue
  • Past issues
  • Specialties
  • Reviews
  • Review series
  • Conversations with Giants in Medicine
  • Video Abstracts
  • In-Press Preview
  • Clinical Research and Public Health
  • Research Letters
  • Letters to the Editor
  • Editorials
  • Commentaries
  • Editor's notes
  • Reviews
  • Viewpoints
  • 100th anniversary
  • Top read articles
  • About
  • Editors
  • Consulting Editors
  • For authors
  • Publication ethics
  • Publication alerts by email
  • Advertising
  • Job board
  • Contact

Issue published May 1, 2014 Previous issue | Next issue

  • Volume 124, Issue 5
Go to section:
  • Reviews
  • Hindsight
  • Commentaries
  • Research Articles
  • Corrigenda

On the cover: Mapping GPCR activation in vivo

Activation of sphingosine-1- phosphate receptor 1 (S1P1; green) is shown in this liver section from SIP1 GFP reporter mice with immunofluorescent labeling of albumin (red) and DAPI staining (blue). On page 2076, Kono et al. report the development of a novel genetic model to uncover S1P1 activation in specific cell types in vivo. Their work not only provides important new insights into S1P1 signaling, but also offers a technique that could be broadly applied to other GPCRs to reveal signaling activity.
Reviews
Uterine NK cells: active regulators at the maternal-fetal interface
Ashley Moffett, Francesco Colucci
Ashley Moffett, Francesco Colucci
Published May 1, 2014
Citation Information: J Clin Invest. 2014;124(5):1872-1879. https://doi.org/10.1172/JCI68107.
View: Text | PDF

Uterine NK cells: active regulators at the maternal-fetal interface

  • Text
  • PDF
Abstract

Pregnancy presents an immunological conundrum because two genetically different individuals coexist. The maternal lymphocytes at the uterine maternal-fetal interface that can recognize mismatched placental cells are T cells and abundant distinctive uterine NK (uNK) cells. Multiple mechanisms exist that avoid damaging T cell responses to the fetus, whereas activation of uNK cells is probably physiological. Indeed, genetic epidemiological data suggest that the variability of NK cell receptors and their MHC ligands define pregnancy success; however, exactly how uNK cells function in normal and pathological pregnancy is still unclear, and any therapies aimed at suppressing NK cells must be viewed with caution. Allorecognition of fetal placental cells by uNK cells is emerging as the key maternal-fetal immune mechanism that regulates placentation.

Authors

Ashley Moffett, Francesco Colucci

×

Emerging therapies for food allergy
Corinne A. Keet, Robert A. Wood
Corinne A. Keet, Robert A. Wood
Published May 1, 2014
Citation Information: J Clin Invest. 2014;124(5):1880-1886. https://doi.org/10.1172/JCI72061.
View: Text | PDF

Emerging therapies for food allergy

  • Text
  • PDF
Abstract

Food allergy is a common condition for which there are currently no approved treatments except avoidance of the allergenic food and treatment of accidental reactions. There are several potential treatments that are under active investigation in animal and human studies, but it is not yet clear what the best approach may be. Here, we review approaches that are currently in clinical trials, including oral, sublingual, and epicutaneous immunotherapy, immunotherapy combined with anti-IgE, and Chinese herbal medicine as well as approaches that are in preclinical or early clinical investigation, including modified protein immunotherapy, adjuvants, DNA vaccines, and helminth administration. We discuss the importance of fully exploring the risks and benefits of any treatment before it is taken to general clinical practice and the need for clarity about the goals of treatment.

Authors

Corinne A. Keet, Robert A. Wood

×
Hindsight
Diabetes, aging, and their tissue complications
Rick Bucala
Rick Bucala
Published May 1, 2014
Citation Information: J Clin Invest. 2014;124(5):1887-1888. https://doi.org/10.1172/JCI75224.
View: Text | PDF

Diabetes, aging, and their tissue complications

  • Text
  • PDF
Abstract

The inactivation of NO by advanced glycation endproducts (AGEs), which accumulate on tissue proteins as a function of age and hyperglycemia, focused attention on the role of these ubiquitous posttranslational modifications in acquired impairments of vascular reactivity and other signaling processes. This observation occurred during a watershed period of basic and translational research in glycation that encompassed new pathologic phenomena and novel intervention strategies. How has the AGE paradigm for the tissue complications of aging and diabetes fared since the identification of the link between these glycation products and NO inactivation, and what lessons may be offered for future investigations?

Authors

Rick Bucala

×
Commentaries
Repurposing staples for viruses: applying peptide design to RSV prophylaxis
Sarah P. Katen, Terence S. Dermody
Sarah P. Katen, Terence S. Dermody
Published April 17, 2014
Citation Information: J Clin Invest. 2014;124(5):1889-1891. https://doi.org/10.1172/JCI75797.
View: Text | PDF

Repurposing staples for viruses: applying peptide design to RSV prophylaxis

  • Text
  • PDF
Abstract

Respiratory syncytial virus (RSV) is responsible for lower respiratory tract infections and annually results in 200,000 deaths worldwide. Despite the burden of RSV-associated disease, treatments and preventative measures are limited. In this issue of JCI, Bird and colleagues describe their work using a peptide stapling technique that allowed synthesis of a stable peptide mimic of a portion of the RSV fusion protein. Pretreatment of cells with the stable peptide effectively blocked virus entry. When introduced into mice prior to RSV exposure, the peptide produced a substantial prophylactic effect. This work provides a new way forward in RSV prevention.

Authors

Sarah P. Katen, Terence S. Dermody

×

A VISTA on PD-1H
Yang Liu
Yang Liu
Published April 17, 2014
Citation Information: J Clin Invest. 2014;124(5):1891-1893. https://doi.org/10.1172/JCI75798.
View: Text | PDF

A VISTA on PD-1H

  • Text
  • PDF
Abstract

Three years ago, two research groups independently identified a previously undescribed T cell cosignaling molecule; one referred to it as V-domain Ig suppressor of T cell activation (VISTA), and the other used the term programmed death-1 homolog (PD-1H). Recombinant and ectopically expressed PD-1H functions as a coinhibitory ligand for T cell responses. However, the function of endogenous PD-1H is not clear. In this issue of the JCI, Flies and colleagues demonstrate that endogenous PD-1H on both T cells and APCs serves as a coinhibitory molecule for T cell activation and provide further support for targeting PD-1H as a therapeutic strategy for transplantation and cancers.

Authors

Yang Liu

×

Cryptococcosis: a model for the understanding of infectious diseases
John R. Perfect
John R. Perfect
Published April 17, 2014
Citation Information: J Clin Invest. 2014;124(5):1893-1895. https://doi.org/10.1172/JCI75241.
View: Text | PDF

Cryptococcosis: a model for the understanding of infectious diseases

  • Text
  • PDF
Abstract

The increase in immunosuppressed patient populations has correlated with a rise in clinical fungal infections, including cryptococcosis. Patient outcome following Cryptococcus infection is linked to initial fungal burden in cerebrospinal fluid (CSF) and fungal clearance following treatment; however, the role of the pathogen in disease prognosis is poorly defined. In this issue of the JCI, Sabiiti and colleagues have directly correlated phenotypic traits of Cryptococcus neoformans with clinical outcome of infected patients. A better understanding of both the host and pathogen contributions to disease etiology will provide more options for targeted treatment strategies.

Authors

John R. Perfect

×

Stargazing microRNA maps a new miR-21 star for cardiac hypertrophy
Ciro Indolfi, Antonio Curcio
Ciro Indolfi, Antonio Curcio
Published April 17, 2014
Citation Information: J Clin Invest. 2014;124(5):1896-1898. https://doi.org/10.1172/JCI75801.
View: Text | PDF

Stargazing microRNA maps a new miR-21 star for cardiac hypertrophy

  • Text
  • PDF
Abstract

Left ventricular hypertrophy is an initial compensatory mechanism in response to cardiac stress that can degenerate into heart failure and sudden cardiac death. Recent studies have shown that microRNAs (miRs) regulate several aspects of cardiovascular diseases. In this issue of the JCI, Bang and colleagues identified an exosome-mediated communication mechanism between cardiac fibroblasts and cardiomyocytes. Specifically, cardiac fibroblasts secrete miR-enriched exosomes, which are subsequently taken up by cardiomyocytes, in which they alter gene expression. In particular, a passenger strand miR, miR-21*, was identified as a potent paracrine factor that induces cardiomyocyte hypertrophy when shuttled through exosomes. These advanced comprehensive analyses represent a major step forward in our understanding of cardiovascular physiopathology, providing a promising adjunctive target for possible therapeutic approaches, namely the miR-mediated paracrine signaling network.

Authors

Ciro Indolfi, Antonio Curcio

×
Research Articles
RIP140 increases APC expression and controls intestinal homeostasis and tumorigenesis
Marion Lapierre, … , Malcolm Parker, Vincent Cavailles
Marion Lapierre, … , Malcolm Parker, Vincent Cavailles
Published March 25, 2014
Citation Information: J Clin Invest. 2014;124(5):1899-1913. https://doi.org/10.1172/JCI65178.
View: Text | PDF

RIP140 increases APC expression and controls intestinal homeostasis and tumorigenesis

  • Text
  • PDF
Abstract

Deregulation of the Wnt/APC/β-catenin signaling pathway is an important consequence of tumor suppressor APC dysfunction. Genetic and molecular data have established that disruption of this pathway contributes to the development of colorectal cancer. Here, we demonstrate that the transcriptional coregulator RIP140 regulates intestinal homeostasis and tumorigenesis. Using Rip140-null mice and mice overexpressing human RIP140, we found that RIP140 inhibited intestinal epithelial cell proliferation and apoptosis. Interestingly, following whole-body irradiation, mice lacking RIP140 exhibited improved regenerative capacity in the intestine, while mice overexpressing RIP140 displayed reduced recovery. Enhanced RIP140 expression strongly repressed human colon cancer cell proliferation in vitro and after grafting onto nude mice. Moreover, in murine tissues and human cancer cells, RIP140 stimulated APC transcription and inhibited β-catenin activation and target gene expression. Finally, RIP140 mRNA and RIP140 protein levels were decreased in human colon cancers compared with those in normal mucosal tissue, and low levels of RIP140 expression in adenocarcinomas from patients correlated with poor prognosis. Together, these results support a tumor suppressor role for RIP140 in colon cancer.

Authors

Marion Lapierre, Sandrine Bonnet, Caroline Bascoul-Mollevi, Imade Ait-Arsa, Stéphan Jalaguier, Maguy Del Rio, Michela Plateroti, Paul Roepman, Marc Ychou, Julie Pannequin, Frédéric Hollande, Malcolm Parker, Vincent Cavailles

×

Autophagy-regulating TP53INP2 mediates muscle wasting and is repressed in diabetes
David Sala, … , Antonio L. Serrano, Antonio Zorzano
David Sala, … , Antonio L. Serrano, Antonio Zorzano
Published April 8, 2014
Citation Information: J Clin Invest. 2014;124(5):1914-1927. https://doi.org/10.1172/JCI72327.
View: Text | PDF

Autophagy-regulating TP53INP2 mediates muscle wasting and is repressed in diabetes

  • Text
  • PDF
Abstract

A precise balance between protein degradation and synthesis is essential to preserve skeletal muscle mass. Here, we found that TP53INP2, a homolog of the Drosophila melanogaster DOR protein that regulates autophagy in cellular models, has a direct impact on skeletal muscle mass in vivo. Using different transgenic mouse models, we demonstrated that muscle-specific overexpression of Tp53inp2 reduced muscle mass, while deletion of Tp53inp2 resulted in muscle hypertrophy. TP53INP2 activated basal autophagy in skeletal muscle and sustained p62-independent autophagic degradation of ubiquitinated proteins. Animals with muscle-specific overexpression of Tp53inp2 exhibited enhanced muscle wasting in streptozotocin-induced diabetes that was dependent on autophagy; however, TP53INP2 ablation mitigated experimental diabetes-associated muscle loss. The overexpression or absence of TP53INP2 did not affect muscle wasting in response to denervation, a condition in which autophagy is blocked, further indicating that TP53INP2 alters muscle mass by activating autophagy. Moreover, TP53INP2 expression was markedly repressed in muscle from patients with type 2 diabetes and in murine models of diabetes. Our results indicate that TP53INP2 negatively regulates skeletal muscle mass through activation of autophagy. Furthermore, we propose that TP53INP2 repression is part of an adaptive mechanism aimed at preserving muscle mass under conditions in which insulin action is deficient.

Authors

David Sala, Saška Ivanova, Natàlia Plana, Vicent Ribas, Jordi Duran, Daniel Bach, Saadet Turkseven, Martine Laville, Hubert Vidal, Monika Karczewska-Kupczewska, Irina Kowalska, Marek Straczkowski, Xavier Testar, Manuel Palacín, Marco Sandri, Antonio L. Serrano, Antonio Zorzano

×

Cytomegalovirus pp65 limits dissemination but is dispensable for persistence
Daniel Malouli, … , Louis J. Picker, Klaus Früh
Daniel Malouli, … , Louis J. Picker, Klaus Früh
Published April 1, 2014
Citation Information: J Clin Invest. 2014;124(5):1928-1944. https://doi.org/10.1172/JCI67420.
View: Text | PDF

Cytomegalovirus pp65 limits dissemination but is dispensable for persistence

  • Text
  • PDF
Abstract

The most abundantly produced virion protein in human cytomegalovirus (HCMV) is the immunodominant phosphoprotein 65 (pp65), which is frequently included in CMV vaccines. Although it is nonessential for in vitro CMV growth, pp65 displays immunomodulatory functions that support a potential role in primary and/or persistent infection. To determine the contribution of pp65 to CMV infection and immunity, we generated a rhesus CMV lacking both pp65 orthologs (RhCMVΔpp65ab). While deletion of pp65ab slightly reduced growth in vitro and increased defective particle formation, the protein composition of secreted virions was largely unchanged. Interestingly, pp65 was not required for primary and persistent infection in animals. Immune responses induced by RhCMVΔpp65ab did not prevent reinfection with rhesus CMV; however, reinfection with RhCMVΔUS2-11, which lacks viral-encoded MHC-I antigen presentation inhibitors, was prevented. Unexpectedly, induction of pp65b-specific T cells alone did not protect against RhCMVΔUS2-11 challenge, suggesting that T cells targeting multiple CMV antigens are required for protection. However, pp65-specific immunity was crucial for controlling viral dissemination during primary infection, as indicated by the marked increase of RhCMVΔpp65ab genome copies in CMV-naive, but not CMV-immune, animals. Our data provide rationale for inclusion of pp65 into CMV vaccines but also demonstrate that pp65-induced T cell responses alone do not recapitulate the protective effect of natural infection.

Authors

Daniel Malouli, Scott G. Hansen, Ernesto S. Nakayasu, Emily E. Marshall, Colette M. Hughes, Abigail B. Ventura, Roxanne M. Gilbride, Matthew S. Lewis, Guangwu Xu, Craig Kreklywich, Nathan Whizin, Miranda Fischer, Alfred W. Legasse, Kasinath Viswanathan, Don Siess, David G. Camp II, Michael K. Axthelm, Christoph Kahl, Victor R. DeFilippis, Richard D. Smith, Daniel N. Streblow, Louis J. Picker, Klaus Früh

×

Methyltransferase G9A regulates T cell differentiation during murine intestinal inflammation
Frann Antignano, … , Megan K. Levings, Colby Zaph
Frann Antignano, … , Megan K. Levings, Colby Zaph
Published March 25, 2014
Citation Information: J Clin Invest. 2014;124(5):1945-1955. https://doi.org/10.1172/JCI69592.
View: Text | PDF

Methyltransferase G9A regulates T cell differentiation during murine intestinal inflammation

  • Text
  • PDF
Abstract

Inflammatory bowel disease (IBD) pathogenesis is associated with dysregulated CD4+ Th cell responses, with intestinal homeostasis depending on the balance between IL-17–producing Th17 and Foxp3+ Tregs. Differentiation of naive T cells into Th17 and Treg subsets is associated with specific gene expression profiles; however, the contribution of epigenetic mechanisms to controlling Th17 and Treg differentiation remains unclear. Using a murine T cell transfer model of colitis, we found that T cell–intrinsic expression of the histone lysine methyltransferase G9A was required for development of pathogenic T cells and intestinal inflammation. G9A-mediated dimethylation of histone H3 lysine 9 (H3K9me2) restricted Th17 and Treg differentiation in vitro and in vivo. H3K9me2 was found at high levels in naive Th cells and was lost following Th cell activation. Loss of G9A in naive T cells was associated with increased chromatin accessibility and heightened sensitivity to TGF-β1. Pharmacological inhibition of G9A methyltransferase activity in WT T cells promoted Th17 and Treg differentiation. Our data indicate that G9A-dependent H3K9me2 is a homeostatic epigenetic checkpoint that regulates Th17 and Treg responses by limiting chromatin accessibility and TGF-β1 responsiveness, suggesting G9A as a therapeutic target for treating intestinal inflammation.

Authors

Frann Antignano, Kyle Burrows, Michael R. Hughes, Jonathan M. Han, Ken J. Kron, Nadia M. Penrod, Menno J. Oudhoff, Steven Kai Hao Wang, Paul H. Min, Matthew J. Gold, Alistair L. Chenery, Mitchell J.S. Braam, Thomas C. Fung, Fabio M.V. Rossi, Kelly M. McNagny, Cheryl H. Arrowsmith, Mathieu Lupien, Megan K. Levings, Colby Zaph

×

Ocular-specific ER stress reduction rescues glaucoma in murine glucocorticoid-induced glaucoma
Gulab S. Zode, … , Abbot F. Clark, Val C. Sheffield
Gulab S. Zode, … , Abbot F. Clark, Val C. Sheffield
Published April 1, 2014
Citation Information: J Clin Invest. 2014;124(5):1956-1965. https://doi.org/10.1172/JCI69774.
View: Text | PDF

Ocular-specific ER stress reduction rescues glaucoma in murine glucocorticoid-induced glaucoma

  • Text
  • PDF
Abstract

Administration of glucocorticoids induces ocular hypertension in some patients. If untreated, these patients can develop a secondary glaucoma that resembles primary open-angle glaucoma (POAG). The underlying pathology of glucocorticoid-induced glaucoma is not fully understood, due in part to lack of an appropriate animal model. Here, we developed a murine model of glucocorticoid-induced glaucoma that exhibits glaucoma features that are observed in patients. Treatment of WT mice with topical ocular 0.1% dexamethasone led to elevation of intraocular pressure (IOP), functional and structural loss of retinal ganglion cells, and axonal degeneration, resembling glucocorticoid-induced glaucoma in human patients. Furthermore, dexamethasone-induced ocular hypertension was associated with chronic ER stress of the trabecular meshwork (TM). Similar to patients, withdrawal of dexamethasone treatment reduced elevated IOP and ER stress in this animal model. Dexamethasone induced the transcriptional factor CHOP, a marker for chronic ER stress, in the anterior segment tissues, and Chop deletion reduced ER stress in these tissues and prevented dexamethasone-induced ocular hypertension. Furthermore, reduction of ER stress in the TM with sodium 4-phenylbutyrate prevented dexamethasone-induced ocular hypertension in WT mice. Our data indicate that ER stress contributes to glucocorticoid-induced ocular hypertension and suggest that reducing ER stress has potential as a therapeutic strategy for treating glucocorticoid-induced glaucoma.

Authors

Gulab S. Zode, Arti B. Sharma, Xiaolei Lin, Charles C. Searby, Kevin Bugge, Gun Hee Kim, Abbot F. Clark, Val C. Sheffield

×

Coinhibitory receptor PD-1H preferentially suppresses CD4+ T cell–mediated immunity
Dallas B. Flies, … , Jessica Jane Ye, Lieping Chen
Dallas B. Flies, … , Jessica Jane Ye, Lieping Chen
Published April 17, 2014
Citation Information: J Clin Invest. 2014;124(5):1966-1975. https://doi.org/10.1172/JCI74589.
View: Text | PDF

Coinhibitory receptor PD-1H preferentially suppresses CD4+ T cell–mediated immunity

  • Text
  • PDF
Abstract

T cell activation is regulated by the interactions of surface receptors with stimulatory and inhibitory ligands. Programmed death-1 homolog (PD-1H, also called VISTA) is a member of the CD28 family of proteins and has been shown to act as a coinhibitory ligand on APCs that suppress T cell responses. Here, we determined that PD-1H functions as a coinhibitory receptor for CD4+ T cells. CD4+ T cells in mice lacking PD-1H exhibited a dramatically increased response to antigen stimulation. Furthermore, delivery of a PD-1H–specific agonist mAb directly inhibited CD4+ T cell activation both in vitro and in vivo, validating a coinhibitory function of PD-1H. In a murine model of acute hepatitis, administration of a PD-1H agonist mAb suppressed CD4+ T cell–mediated acute inflammation. PD-1H–deficient animals were highly resistant to tumor induction in a murine brain glioma model, and depletion of CD4+ T cells, but not CD8+ T cells, promoted tumor formation. Together, our findings suggest that PD-1H has potential as a target of immune modulation in the treatment of human inflammation and malignancies.

Authors

Dallas B. Flies, Xue Han, Tomoe Higuchi, Linghua Zheng, Jingwei Sun, Jessica Jane Ye, Lieping Chen

×

Hepatic nuclear corepressor 1 regulates cholesterol absorption through a TRβ1-governed pathway
Inna Astapova, … , David E. Cohen, Anthony N. Hollenberg
Inna Astapova, … , David E. Cohen, Anthony N. Hollenberg
Published April 8, 2014
Citation Information: J Clin Invest. 2014;124(5):1976-1986. https://doi.org/10.1172/JCI73419.
View: Text | PDF

Hepatic nuclear corepressor 1 regulates cholesterol absorption through a TRβ1-governed pathway

  • Text
  • PDF
Abstract

Transcriptional coregulators are important components of nuclear receptor (NR) signaling machinery and provide additional mechanisms for modulation of NR activity. Expression of a mutated nuclear corepressor 1 (NCoR1) that lacks 2 NR interacting domains (NCoRΔID) in the liver leads to elevated expression of genes regulated by thyroid hormone receptor (TR) and liver X receptor (LXR), both of which control hepatic cholesterol metabolism. Here, we demonstrate that expression of NCoRΔID in mouse liver improves dietary cholesterol tolerance in an LXRα-independent manner. NCoRΔID-associated cholesterol tolerance was primarily due to diminished intestinal cholesterol absorption as the result of changes in the composition and hydrophobicity of the bile salt pool. Alterations of the bile salt pool were mediated by increased expression of genes encoding the bile acid metabolism enzymes CYP27A1 and CYP3A11 as well as canalicular bile salt pump ABCB11. We have determined that these genes are regulated by thyroid hormone and that TRβ1 is recruited to their regulatory regions. Together, these data indicate that interactions between NCoR1 and TR control a specific pathway involved in regulation of cholesterol metabolism and clearance.

Authors

Inna Astapova, Preeti Ramadoss, Ricardo H. Costa-e-Sousa, Felix Ye, Kaila A. Holtz, Yingxia Li, Michele W. Niepel, David E. Cohen, Anthony N. Hollenberg

×

Transporters MCT8 and OATP1C1 maintain murine brain thyroid hormone homeostasis
Steffen Mayerl, … , Theo J. Visser, Heike Heuer
Steffen Mayerl, … , Theo J. Visser, Heike Heuer
Published April 1, 2014
Citation Information: J Clin Invest. 2014;124(5):1987-1999. https://doi.org/10.1172/JCI70324.
View: Text | PDF

Transporters MCT8 and OATP1C1 maintain murine brain thyroid hormone homeostasis

  • Text
  • PDF
Abstract

Allan-Herndon-Dudley syndrome (AHDS), a severe form of psychomotor retardation with abnormal thyroid hormone (TH) parameters, is linked to mutations in the TH-specific monocarboxylate transporter MCT8. In mice, deletion of Mct8 (Mct8 KO) faithfully replicates AHDS-associated endocrine abnormalities; however, unlike patients, these animals do not exhibit neurological impairments. While transport of the active form of TH (T3) across the blood-brain barrier is strongly diminished in Mct8 KO animals, prohormone (T4) can still enter the brain, possibly due to the presence of T4-selective organic anion transporting polypeptide (OATP1C1). Here, we characterized mice deficient for both TH transporters, MCT8 and OATP1C1 (Mct8/Oatp1c1 DKO). Mct8/Oatp1c1 DKO mice exhibited alterations in peripheral TH homeostasis that were similar to those in Mct8 KO mice; however, uptake of both T3 and T4 into the brains of Mct8/Oatp1c1 DKO mice was strongly reduced. Evidence of TH deprivation in the CNS of Mct8/Oatp1c1 DKO mice included highly decreased brain TH content as well as altered deiodinase activities and TH target gene expression. Consistent with delayed cerebellar development and reduced myelination, Mct8/Oatp1c1 DKO mice displayed pronounced locomotor abnormalities. Intriguingly, differentiation of GABAergic interneurons in the cerebral cortex was highly compromised. Our findings underscore the importance of TH transporters for proper brain development and provide a basis to study the pathogenic mechanisms underlying AHDS.

Authors

Steffen Mayerl, Julia Müller, Reinhard Bauer, Sarah Richert, Celia M. Kassmann, Veerle M. Darras, Katrin Buder, Anita Boelen, Theo J. Visser, Heike Heuer

×

Efficient phagocytosis and laccase activity affect the outcome of HIV-associated cryptococcosis
Wilber Sabiiti, … , Robin C. May, Tihana Bicanic
Wilber Sabiiti, … , Robin C. May, Tihana Bicanic
Published April 17, 2014
Citation Information: J Clin Invest. 2014;124(5):2000-2008. https://doi.org/10.1172/JCI72950.
View: Text | PDF Clinical Research and Public Health

Efficient phagocytosis and laccase activity affect the outcome of HIV-associated cryptococcosis

  • Text
  • PDF
Abstract

Background. Cryptococcal meningitis (CM) is a leading cause of HIV-associated mortality globally. High fungal burden in cerebrospinal fluid (CSF) at diagnosis and poor fungal clearance during treatment are recognized adverse prognostic markers; however, the underlying pathogenic factors that drive these clinical manifestations are incompletely understood. We profiled a large set of clinical isolates for established cryptococcal virulence traits to evaluate the contribution of C. neoformans phenotypic diversity to clinical presentation and outcome in human cryptococcosis.

Methods. Sixty-five C. neoformans isolates from clinical trial patients with matched clinical data were assayed in vitro to determine murine macrophage uptake, intracellular proliferation rate (IPR), capsule induction, and laccase activity. Analysis of the correlation between prognostic clinical and host immune parameters and fungal phenotypes was performed using Spearman’s r, while the fungal-dependent impact on long-term survival was determined by Cox regression analysis.

Results. High levels of fungal uptake by macrophages in vitro, but not the IPR, were associated with CSF fungal burden (r = 0.38, P = 0.002) and long-term patient survival (hazard ratio [HR] 2.6, 95% CI 1.2–5.5, P = 0.012). High-uptake strains were hypocapsular (r = –0.28, P = 0.05) and exhibited enhanced laccase activity (r = 0.36, P = 0.003). Fungal isolates with greater laccase activity exhibited heightened survival ex vivo in purified CSF (r = 0.49, P < 0.0001) and resistance to clearance following patient antifungal treatment (r = 0.39, P = 0.003).

Conclusion. These findings underscore the contribution of cryptococcal-phagocyte interactions and laccase-dependent melanin pathways to human clinical presentation and outcome. Furthermore, characterization of fungal-specific pathways that drive clinical manifestation provide potential targets for the development of therapeutics and the management of CM.

Funding. This work was made possible by funding from the Wellcome Trust (WT088148MF), the Medical Research Council (MR/J008176/1), the NIHR Surgical Reconstruction and Microbiology Research Centre and the Lister Institute for Preventive Medicine (to R.C. May), and a Wellcome Trust Intermediate fellowship (089966, to T. Bicanic). The C. neoformans isolates were collected within clinical trials funded by the British Infection Society (fellowship to T. Bicanic), the Wellcome Trust (research training fellowships WT069991, to A.E. Brouwer and WT081794, to J.N. Jarvis), and the Medical Research Council, United Kingdom (76201). The funding sources had no role in the design or conduct of this study, nor in preparation of the manuscript.

Authors

Wilber Sabiiti, Emma Robertson, Mathew A. Beale, Simon A. Johnston, Annemarie E. Brouwer, Angela Loyse, Joseph N. Jarvis, Andrew S. Gilbert, Matthew C. Fisher, Thomas S. Harrison, Robin C. May, Tihana Bicanic

×

CXCL11-dependent induction of FOXP3-negative regulatory T cells suppresses autoimmune encephalomyelitis
Yaniv Zohar, … , Christopher L. Karp, Nathan Karin
Yaniv Zohar, … , Christopher L. Karp, Nathan Karin
Published April 8, 2014
Citation Information: J Clin Invest. 2014;124(5):2009-2022. https://doi.org/10.1172/JCI71951.
View: Text | PDF | Corrigendum | Expression of Concern

CXCL11-dependent induction of FOXP3-negative regulatory T cells suppresses autoimmune encephalomyelitis

  • Text
  • PDF
Abstract

A single G protein–coupled receptor (GPCR) can activate multiple signaling cascades based on the binding of different ligands. The biological relevance of this feature in immune regulation has not been evaluated. The chemokine-binding GPCR CXCR3 is preferentially expressed on CD4+ T cells, and canonically binds 3 structurally related chemokines: CXCL9, CXCL10, and CXCL11. Here we have shown that CXCL10/CXCR3 interactions drive effector Th1 polarization via STAT1, STAT4, and STAT5 phosphorylation, while CXCL11/CXCR3 binding induces an immunotolerizing state that is characterized by IL-10hi (Tr1) and IL-4hi (Th2) cells, mediated via p70 kinase/mTOR in STAT3- and STAT6-dependent pathways. CXCL11 binds CXCR3 with a higher affinity than CXCL10, suggesting that CXCL11 has the potential to restrain inflammatory autoimmunity. We generated a CXCL11-Ig fusion molecule and evaluated its use in the EAE model of inflammatory autoimmune disease. Administration of CXCL11-Ig during the first episode of relapsing EAE in SJL/J mice not only led to rapid remission, but also prevented subsequent relapse. Using GFP-expressing effector CD4+ T cells, we observed that successful therapy was associated with reduced accumulation of these cells at the autoimmune site. Finally, we showed that very low doses of CXCL11 rapidly suppress signs of EAE in C57BL/6 mice lacking functional CXCL11.

Authors

Yaniv Zohar, Gizi Wildbaum, Rostislav Novak, Andrew L. Salzman, Marcus Thelen, Ronen Alon, Yiftah Barsheshet, Christopher L. Karp, Nathan Karin

×

Monocytes expressing CX3CR1 orchestrate the development of vincristine-induced pain
Elizabeth A. Old, … , Mauro Perretti, Marzia Malcangio
Elizabeth A. Old, … , Mauro Perretti, Marzia Malcangio
Published April 17, 2014
Citation Information: J Clin Invest. 2014;124(5):2023-2036. https://doi.org/10.1172/JCI71389.
View: Text | PDF

Monocytes expressing CX3CR1 orchestrate the development of vincristine-induced pain

  • Text
  • PDF
Abstract

A major dose-limiting side effect associated with cancer-treating antineoplastic drugs is the development of neuropathic pain, which is not readily relieved by available analgesics. A better understanding of the mechanisms that underlie pain generation has potential to provide targets for prophylactic management of chemotherapy pain. Here, we delineate a pathway for pain that is induced by the chemotherapeutic drug vincristine sulfate (VCR). In a murine model of chemotherapy-induced allodynia, VCR treatment induced upregulation of endothelial cell adhesion properties, resulting in the infiltration of circulating CX3CR1+ monocytes into the sciatic nerve. At the endothelial-nerve interface, CX3CR1+ monocytes were activated by the chemokine CX3CL1 (also known as fractalkine [FKN]), which promoted production of reactive oxygen species that in turn activated the receptor TRPA1 in sensory neurons and evoked the pain response. Furthermore, mice lacking CX3CR1 exhibited a delay in the development of allodynia following VCR administration. Together, our data suggest that CX3CR1 antagonists and inhibition of FKN proteolytic shedding, possibly by targeting ADAM10/17 and/or cathepsin S, have potential as peripheral approaches for the prophylactic treatment of chemotherapy-induced pain.

Authors

Elizabeth A. Old, Suchita Nadkarni, John Grist, Clive Gentry, Stuart Bevan, Ki-Wook Kim, Adrian J. Mogg, Mauro Perretti, Marzia Malcangio

×

Genetic and pharmacologic inhibition of EPHA2 promotes apoptosis in NSCLC
Katherine R. Amato, … , Nathanael S. Gray, Jin Chen
Katherine R. Amato, … , Nathanael S. Gray, Jin Chen
Published April 8, 2014
Citation Information: J Clin Invest. 2014;124(5):2037-2049. https://doi.org/10.1172/JCI72522.
View: Text | PDF

Genetic and pharmacologic inhibition of EPHA2 promotes apoptosis in NSCLC

  • Text
  • PDF
Abstract

Genome-wide analyses determined previously that the receptor tyrosine kinase (RTK) EPHA2 is commonly overexpressed in non–small cell lung cancers (NSCLCs). EPHA2 overexpression is associated with poor clinical outcomes; therefore, EPHA2 may represent a promising therapeutic target for patients with NSCLC. In support of this hypothesis, here we have shown that targeted disruption of EphA2 in a murine model of aggressive Kras-mutant NSCLC impairs tumor growth. Knockdown of EPHA2 in human NSCLC cell lines reduced cell growth and viability, confirming the epithelial cell autonomous requirements for EPHA2 in NSCLCs. Targeting EPHA2 in NSCLCs decreased S6K1-mediated phosphorylation of cell death agonist BAD and induced apoptosis. Induction of EPHA2 knockdown within established NSCLC tumors in a subcutaneous murine model reduced tumor volume and induced tumor cell death. Furthermore, an ATP-competitive EPHA2 RTK inhibitor, ALW-II-41-27, reduced the number of viable NSCLC cells in a time-dependent and dose-dependent manner in vitro and induced tumor regression in human NSCLC xenografts in vivo. Collectively, these data demonstrate a role for EPHA2 in the maintenance and progression of NSCLCs and provide evidence that ALW-II-41-27 effectively inhibits EPHA2-mediated tumor growth in preclinical models of NSCLC.

Authors

Katherine R. Amato, Shan Wang, Andrew K. Hastings, Victoria M. Youngblood, Pranav R. Santapuram, Haiying Chen, Justin M. Cates, Daniel C. Colvin, Fei Ye, Dana M. Brantley-Sieders, Rebecca S. Cook, Li Tan, Nathanael S. Gray, Jin Chen

×

Intravital imaging of podocyte calcium in glomerular injury and disease
James L. Burford, … , Stuart J. Shankland, János Peti-Peterdi
James L. Burford, … , Stuart J. Shankland, János Peti-Peterdi
Published April 8, 2014
Citation Information: J Clin Invest. 2014;124(5):2050-2058. https://doi.org/10.1172/JCI71702.
View: Text | PDF Technical Advance

Intravital imaging of podocyte calcium in glomerular injury and disease

  • Text
  • PDF
Abstract

Intracellular calcium ([Ca2+]i) signaling mediates physiological and pathological processes in multiple organs, including the renal podocyte; however, in vivo podocyte [Ca2+]i dynamics are not fully understood. Here we developed an imaging approach that uses multiphoton microscopy (MPM) to directly visualize podocyte [Ca2+]i dynamics within the intact kidneys of live mice expressing a fluorescent calcium indicator only in these cells. [Ca2+]i was at a low steady-state level in control podocytes, while Ang II infusion caused a minor elevation. Experimental focal podocyte injury triggered a robust and sustained elevation of podocyte [Ca2+]i around the injury site and promoted cell-to-cell propagating podocyte [Ca2+]i waves along capillary loops. [Ca2+]i wave propagation was ameliorated by inhibitors of purinergic [Ca2+]i signaling as well as in animals lacking the P2Y2 purinergic receptor. Increased podocyte [Ca2+]i resulted in contraction of the glomerular tuft and increased capillary albumin permeability. In preclinical models of renal fibrosis and glomerulosclerosis, high podocyte [Ca2+]i correlated with increased cell motility. Our findings provide a visual demonstration of the in vivo importance of podocyte [Ca2+]i in glomerular pathology and suggest that purinergic [Ca2+]i signaling is a robust and key pathogenic mechanism in podocyte injury. This in vivo imaging approach will allow future detailed investigation of the molecular and cellular mechanisms of glomerular disease in the intact living kidney.

Authors

James L. Burford, Karie Villanueva, Lisa Lam, Anne Riquier-Brison, Matthias J. Hackl, Jeffrey Pippin, Stuart J. Shankland, János Peti-Peterdi

×

Ciliopathy proteins regulate paracrine signaling by modulating proteasomal degradation of mediators
Yangfan P. Liu, … , Brunella Franco, Nicholas Katsanis
Yangfan P. Liu, … , Brunella Franco, Nicholas Katsanis
Published April 1, 2014
Citation Information: J Clin Invest. 2014;124(5):2059-2070. https://doi.org/10.1172/JCI71898.
View: Text | PDF

Ciliopathy proteins regulate paracrine signaling by modulating proteasomal degradation of mediators

  • Text
  • PDF
Abstract

Cilia are critical mediators of paracrine signaling; however, it is unknown whether proteins that contribute to ciliopathies converge on multiple paracrine pathways through a common mechanism. Here, we show that loss of cilopathy-associated proteins Bardet-Biedl syndrome 4 (BBS4) or oral-facial-digital syndrome 1 (OFD1) results in the accumulation of signaling mediators normally targeted for proteasomal degradation. In WT cells, several BBS proteins and OFD1 interacted with proteasomal subunits, and loss of either BBS4 or OFD1 led to depletion of multiple subunits from the centrosomal proteasome. Furthermore, overexpression of proteasomal regulatory components or treatment with proteasomal activators sulforaphane (SFN) and mevalonolactone (MVA) ameliorated signaling defects in cells lacking BBS1, BBS4, and OFD1, in morphant zebrafish embryos, and in induced neurons from Ofd1-deficient mice. Finally, we tested the hypothesis that other proteasome-dependent pathways not known to be associated with ciliopathies are defective in the absence of ciliopathy proteins. We found that loss of BBS1, BBS4, or OFD1 led to decreased NF-κB activity and concomitant IκBβ accumulation and that these defects were ameliorated with SFN treatment. Taken together, our data indicate that basal body proteasomal regulation governs paracrine signaling pathways and suggest that augmenting proteasomal function might benefit ciliopathy patients.

Authors

Yangfan P. Liu, I-Chun Tsai, Manuela Morleo, Edwin C. Oh, Carmen C. Leitch, Filomena Massa, Byung-Hoon Lee, David S. Parker, Daniel Finley, Norann A. Zaghloul, Brunella Franco, Nicholas Katsanis

×

Testicular differentiation factor SF-1 is required for human spleen development
David Zangen, … , Paul Renbaum, Ephrat Levy-Lahad
David Zangen, … , Paul Renbaum, Ephrat Levy-Lahad
Published April 8, 2014
Citation Information: J Clin Invest. 2014;124(5):2071-2075. https://doi.org/10.1172/JCI73186.
View: Text | PDF Brief Report

Testicular differentiation factor SF-1 is required for human spleen development

  • Text
  • PDF
Abstract

The transcription factor steroidogenic factor 1 (SF-1; also known as NR5A1) is a crucial mediator of both steroidogenic and nonsteroidogenic tissue differentiation. Mutations within SF1 underlie different disorders of sexual development (DSD), including sex reversal, spermatogenic failure, ovarian insufficiency, and adrenocortical deficiency. Here, we identified a recessive mutation within SF1 that resulted in a substitution of arginine to glutamine at codon 103 (R103Q) in a child with both severe 46,XY-DSD and asplenia. The R103Q mutation decreased SF-1 transactivation of TLX1, a transcription factor that has been shown to be essential for murine spleen development. Additionally, the SF1 R103Q mutation impaired activation of steroidogenic genes, without affecting synergistic SF-1 and sex-determining region Y (SRY) coactivation of the testis development gene SOX9. Together, our data provide evidence that SF-1 is required for spleen development in humans via transactivation of TLX1 and that mutations that only impair steroidogenesis, without altering the SF1/SRY transactivation of SOX9, can lead to 46,XY-DSD.

Authors

David Zangen, Yotam Kaufman, Ehud Banne, Ariella Weinberg-Shukron, Abdulsalam Abulibdeh, Benjamin P. Garfinkel, Dima Dweik, Moein Kanaan, Núria Camats, Christa Flück, Paul Renbaum, Ephrat Levy-Lahad

×

Sphingosine-1-phosphate receptor 1 reporter mice reveal receptor activation sites in vivo
Mari Kono, … , Ewa M. Turner, Richard L. Proia
Mari Kono, … , Ewa M. Turner, Richard L. Proia
Published March 25, 2014
Citation Information: J Clin Invest. 2014;124(5):2076-2086. https://doi.org/10.1172/JCI71194.
View: Text | PDF Technical Advance

Sphingosine-1-phosphate receptor 1 reporter mice reveal receptor activation sites in vivo

  • Text
  • PDF
Abstract

Activation of the GPCR sphingosine-1-phosphate receptor 1 (S1P1) by sphingosine-1-phosphate (S1P) regulates key physiological processes. S1P1 activation also has been implicated in pathologic processes, including autoimmunity and inflammation; however, the in vivo sites of S1P1 activation under normal and disease conditions are unclear. Here, we describe the development of a mouse model that allows in vivo evaluation of S1P1 activation. These mice, known as S1P1 GFP signaling mice, produce a S1P1 fusion protein containing a transcription factor linked by a protease cleavage site at the C terminus as well as a β-arrestin/protease fusion protein. Activated S1P1 recruits the β-arrestin/protease, resulting in the release of the transcription factor, which stimulates the expression of a GFP reporter gene. Under normal conditions, S1P1 was activated in endothelial cells of lymphoid tissues and in cells in the marginal zone of the spleen, while administration of an S1P1 agonist promoted S1P1 activation in endothelial cells and hepatocytes. In S1P1 GFP signaling mice, LPS-mediated systemic inflammation activated S1P1 in endothelial cells and hepatocytes via hematopoietically derived S1P. These data demonstrate that S1P1 GFP signaling mice can be used to evaluate S1P1 activation and S1P1-active compounds in vivo. Furthermore, this strategy could be potentially applied to any GPCR to identify sites of receptor activation during normal physiology and disease.

Authors

Mari Kono, Ana E. Tucker, Jennifer Tran, Jennifer B. Bergner, Ewa M. Turner, Richard L. Proia

×

Development of a conditionally immortalized human pancreatic β cell line
Raphaël Scharfmann, … , Paul Czernichow, Philippe Ravassard
Raphaël Scharfmann, … , Paul Czernichow, Philippe Ravassard
Published March 25, 2014
Citation Information: J Clin Invest. 2014;124(5):2087-2098. https://doi.org/10.1172/JCI72674.
View: Text | PDF Technical Advance

Development of a conditionally immortalized human pancreatic β cell line

  • Text
  • PDF
Abstract

Diabetic patients exhibit a reduction in β cells, which secrete insulin to help regulate glucose homeostasis; however, little is known about the factors that regulate proliferation of these cells in human pancreas. Access to primary human β cells is limited and a challenge for both functional studies and drug discovery progress. We previously reported the generation of a human β cell line (EndoC-βH1) that was generated from human fetal pancreas by targeted oncogenesis followed by in vivo cell differentiation in mice. EndoC-βH1 cells display many functional properties of adult β cells, including expression of β cell markers and insulin secretion following glucose stimulation; however, unlike primary β cells, EndoC-βH1 cells continuously proliferate. Here, we devised a strategy to generate conditionally immortalized human β cell lines based on Cre-mediated excision of the immortalizing transgenes. The resulting cell line (EndoC-βH2) could be massively amplified in vitro. After expansion, transgenes were efficiently excised upon Cre expression, leading to an arrest of cell proliferation and pronounced enhancement of β cell–specific features such as insulin expression, content, and secretion. Our data indicate that excised EndoC-βH2 cells are highly representative of human β cells and should be a valuable tool for further analysis of human β cells.

Authors

Raphaël Scharfmann, Severine Pechberty, Yasmine Hazhouz, Manon von Bülow, Emilie Bricout-Neveu, Maud Grenier-Godard, Fanny Guez, Latif Rachdi, Matthias Lohmann, Paul Czernichow, Philippe Ravassard

×

Vascular rarefaction mediates whitening of brown fat in obesity
Ippei Shimizu, … , Sonomi Maruyama, Kenneth Walsh
Ippei Shimizu, … , Sonomi Maruyama, Kenneth Walsh
Published April 8, 2014
Citation Information: J Clin Invest. 2014;124(5):2099-2112. https://doi.org/10.1172/JCI71643.
View: Text | PDF

Vascular rarefaction mediates whitening of brown fat in obesity

  • Text
  • PDF
Abstract

Brown adipose tissue (BAT) is a highly vascularized organ with abundant mitochondria that produce heat through uncoupled respiration. Obesity is associated with a reduction of BAT function; however, it is unknown how obesity promotes dysfunctional BAT. Here, using a murine model of diet-induced obesity, we determined that obesity causes capillary rarefaction and functional hypoxia in BAT, leading to a BAT “whitening” phenotype that is characterized by mitochondrial dysfunction, lipid droplet accumulation, and decreased expression of Vegfa. Targeted deletion of Vegfa in adipose tissue of nonobese mice resulted in BAT whitening, supporting a role for decreased vascularity in obesity-associated BAT. Conversely, introduction of VEGF-A specifically into BAT of obese mice restored vascularity, ameliorated brown adipocyte dysfunction, and improved insulin sensitivity. The capillary rarefaction in BAT that was brought about by obesity or Vegfa ablation diminished β-adrenergic signaling, increased mitochondrial ROS production, and promoted mitophagy. These data indicate that overnutrition leads to the development of a hypoxic state in BAT, causing it to whiten through mitochondrial dysfunction and loss. Furthermore, these results link obesity-associated BAT whitening to impaired systemic glucose metabolism.

Authors

Ippei Shimizu, Tamar Aprahamian, Ryosuke Kikuchi, Ayako Shimizu, Kyriakos N. Papanicolaou, Susan MacLauchlan, Sonomi Maruyama, Kenneth Walsh

×

Mucosal delivery of a double-stapled RSV peptide prevents nasopulmonary infection
Gregory H. Bird, … , Shyam S. Mohapatra, Loren D. Walensky
Gregory H. Bird, … , Shyam S. Mohapatra, Loren D. Walensky
Published April 17, 2014
Citation Information: J Clin Invest. 2014;124(5):2113-2124. https://doi.org/10.1172/JCI71856.
View: Text | PDF

Mucosal delivery of a double-stapled RSV peptide prevents nasopulmonary infection

  • Text
  • PDF
Abstract

Respiratory syncytial virus (RSV) infection accounts for approximately 64 million cases of respiratory disease and 200,000 deaths worldwide each year, yet no broadly effective prophylactic or treatment regimen is available. RSV deploys paired, self-associating, heptad repeat domains of its fusion protein, RSV-F, to form a fusogenic 6-helix bundle that enables the virus to penetrate the host cell membrane. Here, we developed hydrocarbon double-stapled RSV fusion peptides that exhibit stabilized α-helical structure and striking proteolytic resistance. Pretreatment with double-stapled RSV peptides that specifically bound to the RSV fusion bundle inhibited infection by both laboratory and clinical RSV isolates in cells and murine infection models. Intranasal delivery of a lead double-stapled RSV peptide effectively prevented viral infection of the nares. A chitosan-based nanoparticle preparation markedly enhanced pulmonary delivery, further preventing progression of RSV infection to the lung. Thus, our results provide a strategy for inhibiting RSV infection by mucosal and endotracheal delivery of double-stapled RSV fusion peptides.

Authors

Gregory H. Bird, Sandhya Boyapalle, Terianne Wong, Kwadwo Opoku-Nsiah, Raminder Bedi, W. Christian Crannell, Alisa F. Perry, Huy Nguyen, Viviana Sampayo, Ankita Devareddy, Subhra Mohapatra, Shyam S. Mohapatra, Loren D. Walensky

×

Senescence-associated SIN3B promotes inflammation and pancreatic cancer progression
Maïté Rielland, … , George Miller, Gregory David
Maïté Rielland, … , George Miller, Gregory David
Published April 1, 2014
Citation Information: J Clin Invest. 2014;124(5):2125-2135. https://doi.org/10.1172/JCI72619.
View: Text | PDF

Senescence-associated SIN3B promotes inflammation and pancreatic cancer progression

  • Text
  • PDF
Abstract

Pancreatic ductal adenocarcinoma (PDAC) is strikingly resistant to conventional therapeutic approaches. We previously demonstrated that the histone deacetylase–associated protein SIN3B is essential for oncogene-induced senescence in cultured cells. Here, using a mouse model of pancreatic cancer, we have demonstrated that SIN3B is required for activated KRAS-induced senescence in vivo. Surprisingly, impaired senescence as the result of genetic inactivation of Sin3B was associated with delayed PDAC progression and correlated with an impaired inflammatory response. In murine and human pancreatic cells and tissues, levels of SIN3B correlated with KRAS-induced production of IL-1α. Furthermore, evaluation of human pancreatic tissue and cancer cells revealed that Sin3B was decreased in control and PDAC samples, compared with samples from patients with pancreatic inflammation. These results indicate that senescence-associated inflammation positively correlates with PDAC progression and suggest that SIN3B has potential as a therapeutic target for inhibiting inflammation-driven tumorigenesis.

Authors

Maïté Rielland, David J. Cantor, Richard Graveline, Cristina Hajdu, Lisa Mara, Beatriz de Diego Diaz, George Miller, Gregory David

×

Cardiac fibroblast–derived microRNA passenger strand-enriched exosomes mediate cardiomyocyte hypertrophy
Claudia Bang, … , Jan Fiedler, Thomas Thum
Claudia Bang, … , Jan Fiedler, Thomas Thum
Published April 17, 2014
Citation Information: J Clin Invest. 2014;124(5):2136-2146. https://doi.org/10.1172/JCI70577.
View: Text | PDF

Cardiac fibroblast–derived microRNA passenger strand-enriched exosomes mediate cardiomyocyte hypertrophy

  • Text
  • PDF
Abstract

In response to stress, the heart undergoes extensive cardiac remodeling that results in cardiac fibrosis and pathological growth of cardiomyocytes (hypertrophy), which contribute to heart failure. Alterations in microRNA (miRNA) levels are associated with dysfunctional gene expression profiles associated with many cardiovascular disease conditions; however, miRNAs have emerged recently as paracrine signaling mediators. Thus, we investigated a potential paracrine miRNA crosstalk between cardiac fibroblasts and cardiomyocytes and found that cardiac fibroblasts secrete miRNA-enriched exosomes. Surprisingly, evaluation of the miRNA content of cardiac fibroblast–derived exosomes revealed a relatively high abundance of many miRNA passenger strands (“star” miRNAs), which normally undergo intracellular degradation. Using confocal imaging and coculture assays, we identified fibroblast exosomal–derived miR-21_3p (miR-21*) as a potent paracrine-acting RNA molecule that induces cardiomyocyte hypertrophy. Proteome profiling identified sorbin and SH3 domain-containing protein 2 (SORBS2) and PDZ and LIM domain 5 (PDLIM5) as miR-21* targets, and silencing SORBS2 or PDLIM5 in cardiomyocytes induced hypertrophy. Pharmacological inhibition of miR-21* in a mouse model of Ang II–induced cardiac hypertrophy attenuated pathology. These findings demonstrate that cardiac fibroblasts secrete star miRNA–enriched exosomes and identify fibroblast-derived miR-21* as a paracrine signaling mediator of cardiomyocyte hypertrophy that has potential as a therapeutic target.

Authors

Claudia Bang, Sandor Batkai, Seema Dangwal, Shashi Kumar Gupta, Ariana Foinquinos, Angelika Holzmann, Annette Just, Janet Remke, Karina Zimmer, Andre Zeug, Evgeni Ponimaskin, Andreas Schmiedl, Xiaoke Yin, Manuel Mayr, Rashi Halder, Andre Fischer, Stefan Engelhardt, Yuanyuan Wei, Andreas Schober, Jan Fiedler, Thomas Thum

×

Melanoma NOS1 expression promotes dysfunctional IFN signaling
Qiuzhen Liu, … , Ena Wang, Francesco M. Marincola
Qiuzhen Liu, … , Ena Wang, Francesco M. Marincola
Published April 1, 2014
Citation Information: J Clin Invest. 2014;124(5):2147-2159. https://doi.org/10.1172/JCI69611.
View: Text | PDF

Melanoma NOS1 expression promotes dysfunctional IFN signaling

  • Text
  • PDF
Abstract

In multiple forms of cancer, constitutive activation of type I IFN signaling is a critical consequence of immune surveillance against cancer; however, PBMCs isolated from cancer patients exhibit depressed STAT1 phosphorylation in response to IFN-α, suggesting IFN signaling dysfunction. Here, we demonstrated in a coculture system that melanoma cells differentially impairs the IFN-α response in PBMCs and that the inhibitory potential of a particular melanoma cell correlates with NOS1 expression. Comparison of gene transcription and array comparative genomic hybridization (aCGH) between melanoma cells from different patients indicated that suppression of IFN-α signaling correlates with an amplification of the NOS1 locus within segment 12q22-24. Evaluation of NOS1 levels in melanomas and IFN responsiveness of purified PBMCs from patients indicated a negative correlation between NOS1 expression in melanomas and the responsiveness of PBMCs to IFN-α. Furthermore, in an explorative study, NOS1 expression in melanoma metastases was negatively associated with patient response to adoptive T cell therapy. This study provides a link between cancer cell phenotype and IFN signal dysfunction in circulating immune cells.

Authors

Qiuzhen Liu, Sara Tomei, Maria Libera Ascierto, Valeria De Giorgi, Davide Bedognetti, Cuilian Dai, Lorenzo Uccellini, Tara Spivey, Zoltan Pos, Jaime Thomas, Jennifer Reinboth, Daniela Murtas, Qianbing Zhang, Lotfi Chouchane, Geoffrey R. Weiss, Craig L. Slingluff Jr., Peter P. Lee, Steven A. Rosenberg, Harvey Alter, Kaitai Yao, Ena Wang, Francesco M. Marincola

×

Platelet-derived S100 family member myeloid-related protein-14 regulates thrombosis
Yunmei Wang, … , Alvin H. Schmaier, Daniel I. Simon
Yunmei Wang, … , Alvin H. Schmaier, Daniel I. Simon
Published April 1, 2014
Citation Information: J Clin Invest. 2014;124(5):2160-2171. https://doi.org/10.1172/JCI70966.
View: Text | PDF

Platelet-derived S100 family member myeloid-related protein-14 regulates thrombosis

  • Text
  • PDF
Abstract

Expression of the gene encoding the S100 calcium–modulated protein family member MRP-14 (also known as S100A9) is elevated in platelets from patients presenting with acute myocardial infarction (MI) compared with those from patients with stable coronary artery disease; however, a causal role for MRP-14 in acute coronary syndromes has not been established. Here, using multiple models of vascular injury, we found that time to arterial thrombotic occlusion was markedly prolonged in Mrp14–/– mice. We observed that MRP-14 and MRP-8/MRP-14 heterodimers (S100A8/A9) are expressed in and secreted by platelets from WT mice and that thrombus formation was reduced in whole blood from Mrp14–/– mice. Infusion of WT platelets, purified MRP-14, or purified MRP-8/MRP-14 heterodimers into Mrp14–/– mice decreased the time to carotid artery occlusion after injury, indicating that platelet-derived MRP-14 directly regulates thrombosis. In contrast, infusion of purified MRP-14 into mice deficient for both MRP-14 and CD36 failed to reduce carotid occlusion times, indicating that CD36 is required for MRP-14–dependent thrombosis. Our data identify a molecular pathway of thrombosis that involves platelet MRP-14 and CD36 and suggest that targeting MRP-14 has potential for treating atherothrombotic disorders, including MI and stroke.

Authors

Yunmei Wang, Chao Fang, Huiyun Gao, Matthew L. Bilodeau, Zijie Zhang, Kevin Croce, Shijian Liu, Toshifumi Morooka, Masashi Sakuma, Kohsuke Nakajima, Shuichi Yoneda, Can Shi, David Zidar, Patrick Andre, Gillian Stephens, Roy L. Silverstein, Nancy Hogg, Alvin H. Schmaier, Daniel I. Simon

×

Excess PLAC8 promotes an unconventional ERK2-dependent EMT in colon cancer
Cunxi Li, … , Lila Solnica-Krezel, Robert J. Coffey
Cunxi Li, … , Lila Solnica-Krezel, Robert J. Coffey
Published April 1, 2014
Citation Information: J Clin Invest. 2014;124(5):2172-2187. https://doi.org/10.1172/JCI71103.
View: Text | PDF

Excess PLAC8 promotes an unconventional ERK2-dependent EMT in colon cancer

  • Text
  • PDF
Abstract

The epithelial-to-mesenchymal transition (EMT) transcriptional program is characterized by repression of E-cadherin (CDH1) and induction of N-cadherin (CDH2), and mesenchymal genes like vimentin (VIM). Placenta-specific 8 (PLAC8) has been implicated in colon cancer; however, how PLAC8 contributes to disease is unknown, and endogenous PLAC8 protein has not been studied. We analyzed zebrafish and human tissues and found that endogenous PLAC8 localizes to the apical domain of differentiated intestinal epithelium. Colon cancer cells with elevated PLAC8 levels exhibited EMT features, including increased expression of VIM and zinc finger E-box binding homeobox 1 (ZEB1), aberrant cell motility, and increased invasiveness. In contrast to classical EMT, PLAC8 overexpression reduced cell surface CDH1 and upregulated P-cadherin (CDH3) without affecting CDH2 expression. PLAC8-induced EMT was linked to increased phosphorylated ERK2 (p-ERK2), and ERK2 knockdown restored cell surface CDH1 and suppressed CDH3, VIM, and ZEB1 upregulation. In vitro, PLAC8 directly bound and inactivated the ERK2 phosphatase DUSP6, thereby increasing p-ERK2. In a murine xenograft model, knockdown of endogenous PLAC8 in colon cancer cells resulted in smaller tumors, reduced local invasion, and decreased p-ERK2. Using MultiOmyx, a multiplex immunofluorescence-based methodology, we observed coexpression of cytosolic PLAC8, CDH3, and VIM at the leading edge of a human colorectal tumor, supporting a role for PLAC8 in cancer invasion in vivo.

Authors

Cunxi Li, Haiting Ma, Yang Wang, Zheng Cao, Ramona Graves-Deal, Anne E. Powell, Alina Starchenko, Gregory D. Ayers, Mary Kay Washington, Vidya Kamath, Keyur Desai, Michael J. Gerdes, Lila Solnica-Krezel, Robert J. Coffey

×

Hydroxycarboxylic acid receptor 2 mediates dimethyl fumarate’s protective effect in EAE
Hui Chen, … , Nina Wettschureck, Markus Schwaninger
Hui Chen, … , Nina Wettschureck, Markus Schwaninger
Published April 1, 2014
Citation Information: J Clin Invest. 2014;124(5):2188-2192. https://doi.org/10.1172/JCI72151.
View: Text | PDF Brief Report

Hydroxycarboxylic acid receptor 2 mediates dimethyl fumarate’s protective effect in EAE

  • Text
  • PDF
Abstract

Taken orally, the drug dimethyl fumarate (DMF) has been shown to improve functional outcomes for patients with MS; however, it is unclear how DMF mediates a protective effect. DMF and, more so, its active metabolite, monomethyl fumarate, are known agonists of the hydroxycarboxylic acid receptor 2 (HCA2), a G protein–coupled membrane receptor. Here, we evaluated the contribution of HCA2 in mediating the protective effect afforded by DMF in EAE, a mouse model of MS. DMF treatment reduced neurological deficit, immune cell infiltration, and demyelination of the spinal cords in wild-type mice, but not in Hca2–/– mice, indicating that HCA2 is required for the therapeutic effect of DMF. In particular, DMF decreased the number of infiltrating neutrophils in a HCA2-dependent manner, likely by interfering with neutrophil adhesion to endothelial cells and chemotaxis. Together, our data indicate that HCA2 mediates the therapeutic effects of DMF in EAE. Furthermore, identification of HCA2 as a molecular target may help to optimize MS therapy.

Authors

Hui Chen, Julian C. Assmann, Antje Krenz, Mahbubur Rahman, Myriam Grimm, Christian M. Karsten, Jörg Köhl, Stefan Offermanns, Nina Wettschureck, Markus Schwaninger

×

Leptin-promoted cilia assembly is critical for normal energy balance
Yu Mi Han, … , Bonghee Lee, Min-Seon Kim
Yu Mi Han, … , Bonghee Lee, Min-Seon Kim
Published March 25, 2014
Citation Information: J Clin Invest. 2014;124(5):2193-2197. https://doi.org/10.1172/JCI69395.
View: Text | PDF Brief Report

Leptin-promoted cilia assembly is critical for normal energy balance

  • Text
  • PDF
Abstract

The majority of mammalian cells have nonmotile primary cilia on their surface that act as antenna-like sensory organelles. Genetic defects that result in ciliary dysfunction are associated with obesity in humans and rodents, which suggests that functional cilia are important for controlling energy balance. Here we demonstrated that neuronal cilia lengths were selectively reduced in hypothalami of obese mice with leptin deficiency and leptin resistance. Treatment of N1 hypothalamic neuron cells with leptin stimulated cilia assembly via inhibition of the tumor suppressors PTEN and glycogen synthase kinase 3β (GSK3β). Induction of short cilia in the hypothalamus of adult mice increased food intake and decreased energy expenditure, leading to a positive energy balance. Moreover, mice with short hypothalamic cilia exhibited attenuated anorectic responses to leptin, insulin, and glucose, which indicates that leptin-induced cilia assembly is essential for sensing these satiety signals by hypothalamic neurons. These data suggest that leptin governs the sensitivity of hypothalamic neurons to metabolic signals by controlling the length of the cell’s antenna.

Authors

Yu Mi Han, Gil Myoung Kang, Kyunghee Byun, Hyuk Wan Ko, Joon Kim, Mi-Seon Shin, Hyun-Kyong Kim, So Young Gil, Ji Hee Yu, Bonghee Lee, Min-Seon Kim

×

Type 1 angiotensin receptors on macrophages ameliorate IL-1 receptor–mediated kidney fibrosis
Jian-dong Zhang, … , Daian Chen, Steven D. Crowley
Jian-dong Zhang, … , Daian Chen, Steven D. Crowley
Published April 17, 2014
Citation Information: J Clin Invest. 2014;124(5):2198-2203. https://doi.org/10.1172/JCI61368.
View: Text | PDF Brief Report

Type 1 angiotensin receptors on macrophages ameliorate IL-1 receptor–mediated kidney fibrosis

  • Text
  • PDF
Abstract

In a wide array of kidney diseases, type 1 angiotensin (AT1) receptors are present on the immune cells that infiltrate the renal interstitium. Here, we examined the actions of AT1 receptors on macrophages in progressive renal fibrosis and found that macrophage-specific AT1 receptor deficiency exacerbates kidney fibrosis induced by unilateral ureteral obstruction (UUO). Macrophages isolated from obstructed kidneys of mice lacking AT1 receptors solely on macrophages had heightened expression of proinflammatory M1 cytokines, including IL-1. Evaluation of isolated AT1 receptor–deficient macrophages confirmed the propensity of these cells to produce exaggerated levels of M1 cytokines, which led to more severe renal epithelial cell damage via IL-1 receptor activation in coculture compared with WT macrophages. A murine kidney crosstransplantation concomitant with UUO model revealed that augmentation of renal fibrosis instigated by AT1 receptor–deficient macrophages is mediated by IL-1 receptor stimulation in the kidney. This study indicates that a key role of AT1 receptors on macrophages is to protect the kidney from fibrosis by limiting activation of IL-1 receptors in the kidney.

Authors

Jian-dong Zhang, Mehul B. Patel, Robert Griffiths, Paul C. Dolber, Phillip Ruiz, Matthew A. Sparks, Johannes Stegbauer, Huixia Jin, Jose A. Gomez, Anne F. Buckley, William S. Lefler, Daian Chen, Steven D. Crowley

×

Laminins affect T cell trafficking and allograft fate
Kristi J. Warren, … , Jonathan S. Bromberg, Bryna E. Burrell
Kristi J. Warren, … , Jonathan S. Bromberg, Bryna E. Burrell
Published April 1, 2014
Citation Information: J Clin Invest. 2014;124(5):2204-2218. https://doi.org/10.1172/JCI73683.
View: Text | PDF

Laminins affect T cell trafficking and allograft fate

  • Text
  • PDF
Abstract

Lymph nodes (LNs) are integral sites for the generation of immune tolerance, migration of CD4+ T cells, and induction of Tregs. Despite the importance of LNs in regulation of inflammatory responses, the LN-specific factors that regulate T cell migration and the precise LN structural domains in which differentiation occurs remain undefined. Using intravital and fluorescent microscopy, we found that alloreactive T cells traffic distinctly into the tolerant LN and colocalize in exclusive regions with alloantigen-presenting cells, a process required for Treg induction. Extracellular matrix proteins, including those of the laminin family, formed regions within the LN that were permissive for colocalization of alloantigen-presenting cells, alloreactive T cells, and Tregs. We identified unique expression patterns of laminin proteins in high endothelial venule basement membranes and the cortical ridge that correlated with alloantigen-specific immunity or immune tolerance. The ratio of laminin α4 to laminin α5 was greater in domains within tolerant LNs, compared with immune LNs, and blocking laminin α4 function or inducing laminin α5 overexpression disrupted T cell and DC localization and transmigration through tolerant LNs. Furthermore, reducing α4 laminin circumvented tolerance induction and induced cardiac allograft inflammation and rejection in murine models. This work identifies laminins as potential targets for immune modulation.

Authors

Kristi J. Warren, Daiki Iwami, Donald G. Harris, Jonathan S. Bromberg, Bryna E. Burrell

×

RSV-encoded NS2 promotes epithelial cell shedding and distal airway obstruction
Rachael M. Liesman, … , Peter L. Collins, Raymond J. Pickles
Rachael M. Liesman, … , Peter L. Collins, Raymond J. Pickles
Published April 8, 2014
Citation Information: J Clin Invest. 2014;124(5):2219-2233. https://doi.org/10.1172/JCI72948.
View: Text | PDF

RSV-encoded NS2 promotes epithelial cell shedding and distal airway obstruction

  • Text
  • PDF
Abstract

Respiratory syncytial virus (RSV) infection is the major cause of bronchiolitis in young children. The factors that contribute to the increased propensity of RSV-induced distal airway disease compared with other commonly encountered respiratory viruses remain unclear. Here, we identified the RSV-encoded nonstructural 2 (NS2) protein as a viral genetic determinant for initiating RSV-induced distal airway obstruction. Infection of human cartilaginous airway epithelium (HAE) and a hamster model of disease with recombinant respiratory viruses revealed that NS2 promotes shedding of infected epithelial cells, resulting in two consequences of virus infection. First, epithelial cell shedding accelerated the reduction of virus titers, presumably by clearing virus-infected cells from airway mucosa. Second, epithelial cells shedding into the narrow-diameter bronchiolar airway lumens resulted in rapid accumulation of detached, pleomorphic epithelial cells, leading to acute distal airway obstruction. Together, these data indicate that RSV infection of the airway epithelium, via the action of NS2, promotes epithelial cell shedding, which not only accelerates viral clearance but also contributes to acute obstruction of the distal airways. Our results identify RSV NS2 as a contributing factor for the enhanced propensity of RSV to cause severe airway disease in young children and suggest NS2 as a potential therapeutic target for reducing the severity of distal airway disease.

Authors

Rachael M. Liesman, Ursula J. Buchholz, Cindy L. Luongo, Lijuan Yang, Alan D. Proia, John P. DeVincenzo, Peter L. Collins, Raymond J. Pickles

×

CaMK4-dependent activation of AKT/mTOR and CREM-α underlies autoimmunity-associated Th17 imbalance
Tomohiro Koga, … , José C. Crispín, George C. Tsokos
Tomohiro Koga, … , José C. Crispín, George C. Tsokos
Published March 25, 2014
Citation Information: J Clin Invest. 2014;124(5):2234-2245. https://doi.org/10.1172/JCI73411.
View: Text | PDF

CaMK4-dependent activation of AKT/mTOR and CREM-α underlies autoimmunity-associated Th17 imbalance

  • Text
  • PDF
Abstract

Tissue inflammation in several autoimmune diseases, including SLE and MS, has been linked to an imbalance of IL-17–producing Th (Th17) cells and Tregs; however, the factors that promote Th17-driven autoimmunity are unclear. Here, we present evidence that the calcium/calmodulin-dependent protein kinase IV (CaMK4) is increased and required during Th17 cell differentiation. Isolation of naive T cells from a murine model of lupus revealed increased levels of CaMK4 following stimulation with Th17-inducing cytokines but not following Treg, Th1, or Th2 induction. Furthermore, naive T cells from mice lacking CaMK4 did not produce IL-17. Genetic or pharmacologic inhibition of CaMK4 decreased the frequency of IL-17–producing T cells and ameliorated EAE and lupus-like disease in murine models. Inhibition of CaMK4 reduced Il17 transcription through decreased activation of the cAMP response element modulator α (CREM-α) and reduced activation of the AKT/mTOR pathway, which is known to enhance Th17 differentiation. Importantly, silencing CaMK4 in T cells from patients with SLE and healthy individuals inhibited Th17 differentiation through reduction of IL17A and IL17F mRNA. Collectively, our results suggest that CaMK4 inhibition has potential as a therapeutic strategy for Th17-driven autoimmune diseases.

Authors

Tomohiro Koga, Christian M. Hedrich, Masayuki Mizui, Nobuya Yoshida, Kotaro Otomo, Linda A. Lieberman, Thomas Rauen, José C. Crispín, George C. Tsokos

×

PD-1 identifies the patient-specific CD8+ tumor-reactive repertoire infiltrating human tumors
Alena Gros, … , James C. Yang, Steven A. Rosenberg
Alena Gros, … , James C. Yang, Steven A. Rosenberg
Published March 25, 2014
Citation Information: J Clin Invest. 2014;124(5):2246-2259. https://doi.org/10.1172/JCI73639.
View: Text | PDF

PD-1 identifies the patient-specific CD8+ tumor-reactive repertoire infiltrating human tumors

  • Text
  • PDF
Abstract

Adoptive transfer of tumor-infiltrating lymphocytes (TILs) can mediate regression of metastatic melanoma; however, TILs are a heterogeneous population, and there are no effective markers to specifically identify and select the repertoire of tumor-reactive and mutation-specific CD8+ lymphocytes. The lack of biomarkers limits the ability to study these cells and develop strategies to enhance clinical efficacy and extend this therapy to other malignancies. Here, we evaluated unique phenotypic traits of CD8+ TILs and TCR β chain (TCRβ) clonotypic frequency in melanoma tumors to identify patient-specific repertoires of tumor-reactive CD8+ lymphocytes. In all 6 tumors studied, expression of the inhibitory receptors programmed cell death 1 (PD-1; also known as CD279), lymphocyte-activation gene 3 (LAG-3; also known as CD223), and T cell immunoglobulin and mucin domain 3 (TIM-3) on CD8+ TILs identified the autologous tumor-reactive repertoire, including mutated neoantigen-specific CD8+ lymphocytes, whereas only a fraction of the tumor-reactive population expressed the costimulatory receptor 4-1BB (also known as CD137). TCRβ deep sequencing revealed oligoclonal expansion of specific TCRβ clonotypes in CD8+PD-1+ compared with CD8+PD-1– TIL populations. Furthermore, the most highly expanded TCRβ clonotypes in the CD8+ and the CD8+PD-1+ populations recognized the autologous tumor and included clonotypes targeting mutated antigens. Thus, in addition to the well-documented negative regulatory role of PD-1 in T cells, our findings demonstrate that PD-1 expression on CD8+ TILs also accurately identifies the repertoire of clonally expanded tumor-reactive cells and reveal a dual importance of PD-1 expression in the tumor microenvironment.

Authors

Alena Gros, Paul F. Robbins, Xin Yao, Yong F. Li, Simon Turcotte, Eric Tran, John R. Wunderlich, Arnold Mixon, Shawn Farid, Mark E. Dudley, Ken-ichi Hanada, Jorge R. Almeida, Sam Darko, Daniel C. Douek, James C. Yang, Steven A. Rosenberg

×

Multifactorial ERβ and NOTCH1 control of squamous differentiation and cancer
Yang Sui Brooks, … , Karine Lefort, G. Paolo Dotto
Yang Sui Brooks, … , Karine Lefort, G. Paolo Dotto
Published April 17, 2014
Citation Information: J Clin Invest. 2014;124(5):2260-2276. https://doi.org/10.1172/JCI72718.
View: Text | PDF

Multifactorial ERβ and NOTCH1 control of squamous differentiation and cancer

  • Text
  • PDF
Abstract

Downmodulation or loss-of-function mutations of the gene encoding NOTCH1 are associated with dysfunctional squamous cell differentiation and development of squamous cell carcinoma (SCC) in skin and internal organs. While NOTCH1 receptor activation has been well characterized, little is known about how NOTCH1 gene transcription is regulated. Using bioinformatics and functional screening approaches, we identified several regulators of the NOTCH1 gene in keratinocytes, with the transcription factors DLX5 and EGR3 and estrogen receptor β (ERβ) directly controlling its expression in differentiation. DLX5 and ERG3 are required for RNA polymerase II (PolII) recruitment to the NOTCH1 locus, while ERβ controls NOTCH1 transcription through RNA PolII pause release. Expression of several identified NOTCH1 regulators, including ERβ, is frequently compromised in skin, head and neck, and lung SCCs and SCC-derived cell lines. Furthermore, a keratinocyte ERβ–dependent program of gene expression is subverted in SCCs from various body sites, and there are consistent differences in mutation and gene-expression signatures of head and neck and lung SCCs in female versus male patients. Experimentally increased ERβ expression or treatment with ERβ agonists inhibited proliferation of SCC cells and promoted NOTCH1 expression and squamous differentiation both in vitro and in mouse xenotransplants. Our data identify a link between transcriptional control of NOTCH1 expression and the estrogen response in keratinocytes, with implications for differentiation therapy of squamous cancer.

Authors

Yang Sui Brooks, Paola Ostano, Seung-Hee Jo, Jun Dai, Spiro Getsios, Piotr Dziunycz, Günther F.L. Hofbauer, Kara Cerveny, Giovanna Chiorino, Karine Lefort, G. Paolo Dotto

×

Apolipoprotein O is mitochondrial and promotes lipotoxicity in heart
Annie Turkieh, … , Philippe Rouet, Fatima Smih
Annie Turkieh, … , Philippe Rouet, Fatima Smih
Published April 17, 2014
Citation Information: J Clin Invest. 2014;124(5):2277-2286. https://doi.org/10.1172/JCI74668.
View: Text | PDF

Apolipoprotein O is mitochondrial and promotes lipotoxicity in heart

  • Text
  • PDF
Abstract

Diabetic cardiomyopathy is a secondary complication of diabetes with an unclear etiology. Based on a functional genomic evaluation of obesity-associated cardiac gene expression, we previously identified and cloned the gene encoding apolipoprotein O (APOO), which is overexpressed in hearts from diabetic patients. Here, we generated APOO-Tg mice, transgenic mouse lines that expresses physiological levels of human APOO in heart tissue. APOO-Tg mice fed a high-fat diet exhibited depressed ventricular function with reduced fractional shortening and ejection fraction, and myocardial sections from APOO-Tg mice revealed mitochondrial degenerative changes. In vivo fluorescent labeling and subcellular fractionation revealed that APOO localizes with mitochondria. Furthermore, APOO enhanced mitochondrial uncoupling and respiration, both of which were reduced by deletion of the N-terminus and by targeted knockdown of APOO. Consequently, fatty acid metabolism and ROS production were enhanced, leading to increased AMPK phosphorylation and Ppara and Pgc1a expression. Finally, we demonstrated that the APOO-induced cascade of events generates a mitochondrial metabolic sink whereby accumulation of lipotoxic byproducts leads to lipoapoptosis, loss of cardiac cells, and cardiomyopathy, mimicking the diabetic heart–associated metabolic phenotypes. Our data suggest that APOO represents a link between impaired mitochondrial function and cardiomyopathy onset, and targeting APOO-dependent metabolic remodeling has potential as a strategy to adjust heart metabolism and protect the myocardium from impaired contractility.

Authors

Annie Turkieh, Céline Caubère, Manon Barutaut, Franck Desmoulin, Romain Harmancey, Michel Galinier, Matthieu Berry, Camille Dambrin, Carlo Polidori, Louis Casteilla, François Koukoui, Philippe Rouet, Fatima Smih

×
Corrigenda
Dapagliflozin improves muscle insulin sensitivity but enhances endogenous glucose production
Aurora Merovci, … , Muhammad A. Abdul-Ghani, Ralph A. DeFronzo
Aurora Merovci, … , Muhammad A. Abdul-Ghani, Ralph A. DeFronzo
Published May 1, 2014
Citation Information: J Clin Invest. 2014;124(5):2287-2287. https://doi.org/10.1172/JCI76184.
View: Text | PDF | Amended Article

Dapagliflozin improves muscle insulin sensitivity but enhances endogenous glucose production

  • Text
  • PDF
Abstract

Authors

Aurora Merovci, Carolina Solis-Herrera, Giuseppe Daniele, Roy Eldor, Teresa Vanessa Fiorentino, Devjit Tripathy, Juan Xiong, Zandra Perez, Luke Norton, Muhammad A. Abdul-Ghani, Ralph A. DeFronzo

×

Cell-specific translational profiling in acute kidney injury
Jing Liu, … , Winston A. Hide, Andrew P. McMahon
Jing Liu, … , Winston A. Hide, Andrew P. McMahon
Published May 1, 2014
Citation Information: J Clin Invest. 2014;124(5):2288-2288. https://doi.org/10.1172/JCI76261.
View: Text | PDF | Amended Article

Cell-specific translational profiling in acute kidney injury

  • Text
  • PDF
Abstract

Authors

Jing Liu, A. Michaela Krautzberger, Shannan H. Sui, Oliver M. Hofmann, Ying Chen, Manfred Baetscher, Ivica Grgic, Sanjeev Kumar, Benjamin D. Humphreys, Winston A. Hide, Andrew P. McMahon

×
Advertisement

Copyright © 2025 American Society for Clinical Investigation
ISSN: 0021-9738 (print), 1558-8238 (online)

Sign up for email alerts