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 February 15, 2024 Previous issue | Next issue

  • Volume 134, Issue 4
Go to section:
  • 100th Anniversary Viewpoints
  • Viewpoint
  • Review Series
  • Editor's note
  • Commentaries
  • Research Letter
  • Research Articles
  • Corrigendum

On the cover: p53 loss drives lipid droplet growth

Xu et al. report that loss of p53 leads to dysregulation of choline metabolism and lipid droplet growth and couples perturbed lipid droplet homeostasis to tumorigenesis. The cover image shows lipid droplet (green) morphology in p53 WT HepG2 cells. Image credit: Xiuduan Xu.

100th Anniversary Viewpoints
Nitric oxide in vascular biology: elegance in complexity
Joseph Loscalzo
Joseph Loscalzo
Published February 15, 2024
Citation Information: J Clin Invest. 2024;134(4):e176747. https://doi.org/10.1172/JCI176747.
View: Text | PDF

Nitric oxide in vascular biology: elegance in complexity

  • Text
  • PDF
Abstract

Authors

Joseph Loscalzo

×

How the JCI’s most-cited paper sparked the field of lipoprotein research
Michael S. Brown, Joseph L. Goldstein
Michael S. Brown, Joseph L. Goldstein
Published February 15, 2024
Citation Information: J Clin Invest. 2024;134(4):e177475. https://doi.org/10.1172/JCI177475.
View: Text | PDF

How the JCI’s most-cited paper sparked the field of lipoprotein research

  • Text
  • PDF
Abstract

Authors

Michael S. Brown, Joseph L. Goldstein

×
Viewpoint
Status check: next-generation sequencing for infectious-disease diagnostics
Kyle G. Rodino, Patricia J. Simner
Kyle G. Rodino, Patricia J. Simner
Published February 15, 2024
Citation Information: J Clin Invest. 2024;134(4):e178003. https://doi.org/10.1172/JCI178003.
View: Text | PDF

Status check: next-generation sequencing for infectious-disease diagnostics

  • Text
  • PDF
Abstract

Authors

Kyle G. Rodino, Patricia J. Simner

×
Review Series
Hereditary hemorrhagic telangiectasia: from signaling insights to therapeutic advances
Tala Al Tabosh, … , Sophie Dupuis-Girod, Sabine Bailly
Tala Al Tabosh, … , Sophie Dupuis-Girod, Sabine Bailly
Published February 15, 2024
Citation Information: J Clin Invest. 2024;134(4):e176379. https://doi.org/10.1172/JCI176379.
View: Text | PDF

Hereditary hemorrhagic telangiectasia: from signaling insights to therapeutic advances

  • Text
  • PDF
Abstract

Hereditary hemorrhagic telangiectsia (HHT) is an inherited vascular disorder with highly variable expressivity, affecting up to 1 in 5,000 individuals. This disease is characterized by small arteriovenous malformations (AVMs) in mucocutaneous areas (telangiectases) and larger visceral AVMs in the lungs, liver, and brain. HHT is caused by loss-of-function mutations in the BMP9-10/ENG/ALK1/SMAD4 signaling pathway. This Review presents up-to-date insights on this mutated signaling pathway and its crosstalk with proangiogenic pathways, in particular the VEGF pathway, that has allowed the repurposing of new drugs for HHT treatment. However, despite the substantial benefits of these new treatments in terms of alleviating symptom severity, this not-so-uncommon bleeding disorder still currently lacks any FDA- or European Medicines Agency–approved (EMA-approved) therapies.

Authors

Tala Al Tabosh, Mohammad Al Tarrass, Laura Tourvieilhe, Alexandre Guilhem, Sophie Dupuis-Girod, Sabine Bailly

×
Editor's note
Cytoplasmic transcription factor sequestration drives the pathogenesis of a rearranged leukemia
Ali Shilatifard
Ali Shilatifard
Published February 15, 2024
Citation Information: J Clin Invest. 2024;134(4):e179105. https://doi.org/10.1172/JCI179105.
View: Text | PDF

Cytoplasmic transcription factor sequestration drives the pathogenesis of a rearranged leukemia

  • Text
  • PDF
Abstract

Authors

Ali Shilatifard

×
Commentaries
Lipid droplets in the endothelium: The missing link between metabolic syndrome and cardiovascular disease?
Iris Z. Jaffe, S. Ananth Karumanchi
Iris Z. Jaffe, S. Ananth Karumanchi
Published February 15, 2024
Citation Information: J Clin Invest. 2024;134(4):e176347. https://doi.org/10.1172/JCI176347.
View: Text | PDF

Lipid droplets in the endothelium: The missing link between metabolic syndrome and cardiovascular disease?

  • Text
  • PDF
Abstract

The physiology of lipid droplets (LDs) has been most extensively characterized in adipocytes, but LDs also accumulate in endothelial cells lining blood vessels in response to changing levels of triglycerides. In recent issues of the JCI, two independent papers highlight a direct role of endothelial LDs in the genesis of hypertension and atherosclerosis in rodent models. Kim et al. demonstrated that accumulation of LDs in the endothelium leads to hypertension, impairs endothelial function, and accelerates atherosclerosis. Boutagy, Gamez-Mendez, et al. knocked out Atgl in the endothelium and confirmed triglyceride accumulation in endothelial cells that was associated with reduced NO synthesis and impaired endothelial-dependent vasodilation. These data suggest that enhancing triglyceride breakdown in the endothelium could provide a treatment target for patients with metabolic syndrome.

Authors

Iris Z. Jaffe, S. Ananth Karumanchi

×

Yoda1 opens the lymphatic path for craniosynostosis therapy
Aleksanteri Aspelund, Kari Alitalo
Aleksanteri Aspelund, Kari Alitalo
Published February 15, 2024
Citation Information: J Clin Invest. 2024;134(4):e176858. https://doi.org/10.1172/JCI176858.
View: Text | PDF

Yoda1 opens the lymphatic path for craniosynostosis therapy

  • Text
  • PDF
Abstract

The rediscovery of meningeal lymphatic vessels (MLVs) has sparked research interest in their function in numerous neurological pathologies. Craniosynostosis (CS) is caused by a premature fusion of cranial sutures during development. In this issue of the JCI, Matrongolo and colleagues show that Twist1-haploinsufficient mice that develop CS exhibit raised intracranial pressure, diminished cerebrospinal fluid (CSF) outflow, and impaired paravascular CSF-brain flow; all features that were associated with MLV defects and exacerbated pathology in mouse models of Alzheimer’s disease. Activation of the mechanosensor Piezo1 with Yoda1 restored MLV function and CSF perfusion in CS models and in aged mice, opening an avenue for further development of therapeutics.

Authors

Aleksanteri Aspelund, Kari Alitalo

×

NKT cells in the antitumor response: the β version?
Mitchell Kronenberg, Isaac Engel
Mitchell Kronenberg, Isaac Engel
Published February 15, 2024
Citation Information: J Clin Invest. 2024;134(4):e177663. https://doi.org/10.1172/JCI177663.
View: Text | PDF

NKT cells in the antitumor response: the β version?

  • Text
  • PDF
Abstract

NKT cells recognize glycolipids presented by CD1d-expressing antigen-presenting cells (APCs) and include type I NKT cells with antitumor function and type II NKT cells, which have been reported to suppress the antitumor response. Some type II NKT cells recognize sulfatide, a glycosphingolipid with a sulfate modification of the sugar. Type I NKT cells recognize different glycosphingolipids. In this issue of the JCI, Nishio and colleagues showed that APCs could process sulfatide antigens, analogous to protein processing for peptide-reactive T cells. Antigen processing in lysosomes removed sulfate to generate a glycosphingolipid that stimulated type I NKT cells and thereby turned an antigen with no antitumor activity into one that not only stimulated type I NKT cells but also stimulated antitumor responses. These findings may extend to the development of glycolipid antigens that could stimulate anticancer responses via antigen processing by APCs.

Authors

Mitchell Kronenberg, Isaac Engel

×

Reduced Bik expression drives low-grade airway inflammation and increased risk for COPD in females
Irina Petrache, David W.H. Riches
Irina Petrache, David W.H. Riches
Published February 15, 2024
Citation Information: J Clin Invest. 2024;134(4):e177753. https://doi.org/10.1172/JCI177753.
View: Text | PDF

Reduced Bik expression drives low-grade airway inflammation and increased risk for COPD in females

  • Text
  • PDF
Abstract

Chronic low-grade inflammation is increasingly recognized as a subtle yet potent risk factor for a multitude of age-related disorders, including respiratory diseases, cardiovascular conditions, metabolic syndromes, autoimmunity, and cancer. In this issue of the JCI, Mebratu, Jones, and colleagues shed new light on the mechanisms that promote low-grade airway inflammation and how this contributes to the development of chronic obstructive pulmonary disease (COPD). Their finding that Bik deficiency leads to spontaneous emphysema in female mice, but not in males, marks a notable advancement in our understanding of how inflammatory processes can diverge based on biological sex. This finding is of clinical relevance, given the vulnerability of women to developing COPD.

Authors

Irina Petrache, David W.H. Riches

×

Targeting de novo lipogenesis to mitigate kidney disease
Haikuo Li, Benjamin D. Humphreys
Haikuo Li, Benjamin D. Humphreys
Published February 15, 2024
Citation Information: J Clin Invest. 2024;134(4):e178125. https://doi.org/10.1172/JCI178125.
View: Text | PDF

Targeting de novo lipogenesis to mitigate kidney disease

  • Text
  • PDF
Abstract

Ten percent of the population worldwide suffers from chronic kidney disease (CKD), but the mechanisms driving CKD pathology are incompletely understood. While dysregulated lipid metabolism is one hallmark of CKD, the pathogenesis of cellular lipid accumulation remains unclear. In this issue of the JCI, Mukhi et al. Identify acyl-CoA synthetase short-chain family 2 (ACSS2) as a disease risk gene and demonstrate a role for ACSS2 in de novo lipogenesis (DNL). Notably, genetic or pharmacological inhibition of DNL protected against kidney disease progression in mice. These findings warrant evaluation of DNL inhibition with respect to efficacy and safety in people with CKD.

Authors

Haikuo Li, Benjamin D. Humphreys

×
Research Letter
Cancer-associated fibroblast–secreted collagen is associated with immune inhibitor receptor LAIR1 in gliomas
Shashwat Tripathi, … , Michael DeCuypere, Amy B. Heimberger
Shashwat Tripathi, … , Michael DeCuypere, Amy B. Heimberger
Published February 15, 2024
Citation Information: J Clin Invest. 2024;134(4):e176613. https://doi.org/10.1172/JCI176613.
View: Text | PDF

Cancer-associated fibroblast–secreted collagen is associated with immune inhibitor receptor LAIR1 in gliomas

  • Text
  • PDF
Abstract

Authors

Shashwat Tripathi, Hinda Najem, Corey Dussold, Sebastian Pacheco, Jason Miska, Kathleen McCortney, Alicia Steffens, Jordain Walshon, Daniel Winkowski, Michael Cloney, Matthew Ordon, William Gibson, Hanna Kemeny, Mark Youngblood, Rebecca Du, James Mossner, Pavlos Texakalidis, Annelise Sprau, Matthew Tate, Charles David James, Craig M. Horbinski, Nitin R. Wadhwani, Maciej S. Lesniak, Sandi Lam, Ankita Sati, Manish Aghi, Michael DeCuypere, Amy B. Heimberger

×
Research Articles
Heterozygous mutations in the C-terminal domain of COPA underlie a complex autoinflammatory syndrome
Selket Delafontaine, … , Jérôme Delon, Isabelle Meyts
Selket Delafontaine, … , Jérôme Delon, Isabelle Meyts
Published January 4, 2024
Citation Information: J Clin Invest. 2024;134(4):e163604. https://doi.org/10.1172/JCI163604.
View: Text | PDF

Heterozygous mutations in the C-terminal domain of COPA underlie a complex autoinflammatory syndrome

  • Text
  • PDF
Abstract

Mutations in the N-terminal WD40 domain of coatomer protein complex subunit α (COPA) cause a type I interferonopathy, typically characterized by alveolar hemorrhage, arthritis, and nephritis. We described 3 heterozygous mutations in the C-terminal domain (CTD) of COPA (p.C1013S, p.R1058C, and p.R1142X) in 6 children from 3 unrelated families with a similar syndrome of autoinflammation and autoimmunity. We showed that these CTD COPA mutations disrupt the integrity and the function of coat protein complex I (COPI). In COPAR1142X and COPAR1058C fibroblasts, we demonstrated that COPI dysfunction causes both an anterograde ER-to-Golgi and a retrograde Golgi-to-ER trafficking defect. The disturbed intracellular trafficking resulted in a cGAS/STING-dependent upregulation of the type I IFN signaling in patients and patient-derived cell lines, albeit through a distinct molecular mechanism in comparison with mutations in the WD40 domain of COPA. We showed that CTD COPA mutations induce an activation of ER stress and NF-κB signaling in patient-derived primary cell lines. These results demonstrate the importance of the integrity of the CTD of COPA for COPI function and homeostatic intracellular trafficking, essential to ER homeostasis. CTD COPA mutations result in disease by increased ER stress, disturbed intracellular transport, and increased proinflammatory signaling.

Authors

Selket Delafontaine, Alberto Iannuzzo, Tarin M. Bigley, Bram Mylemans, Ruchit Rana, Pieter Baatsen, Maria Cecilia Poli, Daisy Rymen, Katrien Jansen, Djalila Mekahli, Ingele Casteels, Catherine Cassiman, Philippe Demaerel, Alice Lepelley, Marie-Louise Frémond, Rik Schrijvers, Xavier Bossuyt, Katlijn Vints, Wim Huybrechts, Rachida Tacine, Karen Willekens, Anniek Corveleyn, Bram Boeckx, Marco Baggio, Lisa Ehlers, Sebastian Munck, Diether Lambrechts, Arnout Voet, Leen Moens, Giorgia Bucciol, Megan A. Cooper, Carla M. Davis, Jérôme Delon, Isabelle Meyts

×

Lysosomal processing of sulfatide analogs alters target NKT cell specificity and immune responses in cancer
Kumiko Nishio, … , Amy R. Howell, Jay A. Berzofsky
Kumiko Nishio, … , Amy R. Howell, Jay A. Berzofsky
Published December 21, 2023
Citation Information: J Clin Invest. 2024;134(4):e165281. https://doi.org/10.1172/JCI165281.
View: Text | PDF

Lysosomal processing of sulfatide analogs alters target NKT cell specificity and immune responses in cancer

  • Text
  • PDF
Abstract

In a structure-function study of sulfatides that typically stimulate type II NKT cells, we made an unexpected discovery. We compared analogs with sphingosine or phytosphingosine chains and 24-carbon acyl chains with 0-1-2 double bonds (C or pC24:0, 24:1, or 24:2). C24:1 and C24:2 sulfatide presented by the CD1d monomer on plastic stimulated type II, not type I, NKT cell hybridomas, as expected. Unexpectedly, when presented by bone marrow–derived DCs (BMDCs), C24:2 reversed specificity to stimulate type I, not type II, NKT cell hybridomas, mimicking the corresponding β-galactosylceramide (βGalCer) without sulfate. C24:2 induced IFN-γ–dependent immunoprotection against CT26 colon cancer lung metastases, skewed the cytokine profile, and activated conventional DC subset 1 cells (cDC1s). This was abrogated by blocking lysosomal processing with bafilomycin A1, or by sulfite blocking of arylsulfatase or deletion of this enyzme that cleaves off sulfate. Thus, C24:2 was unexpectedly processed in BMDCs from a type II to a type I NKT cell–stimulating ligand, promoting tumor immunity. We believe this is the first discovery showing that antigen processing of glycosylceramides alters the specificity for the target cell, reversing the glycolipid’s function from stimulating type II NKT cells to stimulating type I NKT cells, thereby introducing protective functional activity in cancer. We also believe our study uncovers a new role for antigen processing that does not involve MHC loading but rather alteration of which type of cell is responding.

Authors

Kumiko Nishio, Lise Pasquet, Kaddy Camara, Julia DiSapio, Kevin S. Hsu, Shingo Kato, Anja Bloom, Stewart K. Richardson, Joshua A. Welsh, Tianbo Jiang, Jennifer C. Jones, Susanna Cardell, Hiroshi Watarai, Masaki Terabe, Purevdorj B. Olkhanud, Amy R. Howell, Jay A. Berzofsky

×

A human obesity-associated MC4R mutation with defective Gq/11α signaling leads to hyperphagia in mice
Peter J. Metzger, … , Min Chen, Lee S. Weinstein
Peter J. Metzger, … , Min Chen, Lee S. Weinstein
Published January 4, 2024
Citation Information: J Clin Invest. 2024;134(4):e165418. https://doi.org/10.1172/JCI165418.
View: Text | PDF

A human obesity-associated MC4R mutation with defective Gq/11α signaling leads to hyperphagia in mice

  • Text
  • PDF
Abstract

Melanocortin 4 receptor (MC4R) mutations are the most common cause of human monogenic obesity and are associated with hyperphagia and increased linear growth. While MC4R is known to activate Gsα/cAMP signaling, a substantial proportion of obesity-associated MC4R mutations do not affect MC4R/Gsα signaling. To further explore the role of specific MC4R signaling pathways in the regulation of energy balance, we examined the signaling properties of one such mutant, MC4R (F51L), as well as the metabolic consequences of MC4RF51L mutation in mice. The MC4RF51L mutation produced a specific defect in MC4R/Gq/11α signaling and led to obesity, hyperphagia, and increased linear growth in mice. The ability of a melanocortin agonist to acutely inhibit food intake when delivered to the paraventricular nucleus (PVN) was lost in MC4RF51L mice, as well as in WT mice in which a specific Gq/11α inhibitor was delivered to the PVN; this provided evidence that a Gsα-independent signaling pathway, namely Gq/11α, significantly contributes to the actions of MC4R on food intake and linear growth. These results suggest that a biased MC4R agonist that primarily activates Gq/11α may be a potential agent to treat obesity with limited untoward cardiovascular and other side effects.

Authors

Peter J. Metzger, Aileen Zhang, Bradley A. Carlson, Hui Sun, Zhenzhong Cui, Yongqi Li, Marshal T. Jahnke, Daniel R. Layton, Meenakshi B. Gupta, Naili Liu, Evi Kostenis, Oksana Gavrilova, Min Chen, Lee S. Weinstein

×

Microbiota-dependent indole production stimulates the development of collagen-induced arthritis in mice
Brenda J. Seymour, … , Sean P. Colgan, Kristine A. Kuhn
Brenda J. Seymour, … , Sean P. Colgan, Kristine A. Kuhn
Published December 19, 2023
Citation Information: J Clin Invest. 2024;134(4):e167671. https://doi.org/10.1172/JCI167671.
View: Text | PDF

Microbiota-dependent indole production stimulates the development of collagen-induced arthritis in mice

  • Text
  • PDF
Abstract

Altered tryptophan catabolism has been identified in inflammatory diseases like rheumatoid arthritis (RA) and spondyloarthritis (SpA), but the causal mechanisms linking tryptophan metabolites to disease are unknown. Using the collagen-induced arthritis (CIA) model, we identified alterations in tryptophan metabolism, and specifically indole, that correlated with disease. We demonstrated that both bacteria and dietary tryptophan were required for disease and that indole supplementation was sufficient to induce disease in their absence. When mice with CIA on a low-tryptophan diet were supplemented with indole, we observed significant increases in serum IL-6, TNF, and IL-1β; splenic RORγt+CD4+ T cells and ex vivo collagen-stimulated IL-17 production; and a pattern of anti-collagen antibody isotype switching and glycosylation that corresponded with increased complement fixation. IL-23 neutralization reduced disease severity in indole-induced CIA. Finally, exposure of human colonic lymphocytes to indole increased the expression of genes involved in IL-17 signaling and plasma cell activation. Altogether, we propose a mechanism by which intestinal dysbiosis during inflammatory arthritis results in altered tryptophan catabolism, leading to indole stimulation of arthritis development. Blockade of indole generation may present a unique therapeutic pathway for RA and SpA.

Authors

Brenda J. Seymour, Brandon Trent, Brendan E. Allen, Adam J. Berlinberg, Jimmy Tangchittsumran, Widian K. Jubair, Meagan E. Chriswell, Sucai Liu, Alfredo Ornelas, Andrew Stahly, Erica E. Alexeev, Alexander S. Dowdell, Sunny L. Sneed, Sabrina Fechtner, Jennifer M. Kofonow, Charles E. Robertson, Stephanie M. Dillon, Cara C. Wilson, Robert M. Anthony, Daniel N. Frank, Sean P. Colgan, Kristine A. Kuhn

×

CTRP4/interleukin-6 receptor signaling ameliorates autoimmune encephalomyelitis by suppressing Th17 cell differentiation
Lulu Cao, … , Xiaoxin Zhu, Lu Wang
Lulu Cao, … , Xiaoxin Zhu, Lu Wang
Published November 28, 2023
Citation Information: J Clin Invest. 2024;134(4):e168384. https://doi.org/10.1172/JCI168384.
View: Text | PDF

CTRP4/interleukin-6 receptor signaling ameliorates autoimmune encephalomyelitis by suppressing Th17 cell differentiation

  • Text
  • PDF
Abstract

C1q/TNF-related protein 4 (CTRP4) is generally thought to be released extracellularly and plays a critical role in energy metabolism and protecting against sepsis. However, its physiological functions in autoimmune diseases have not been thoroughly explored. In this study, we demonstrate that Th17 cell–associated experimental autoimmune encephalomyelitis was greatly exacerbated in Ctrp4–/– mice compared with WT mice due to increased Th17 cell infiltration. The absence of Ctrp4 promoted the differentiation of naive CD4+ T cells into Th17 cells in vitro. Mechanistically, CTRP4 interfered with the interaction between IL-6 and the IL-6 receptor (IL-6R) by directly competing to bind with IL-6R, leading to suppression of IL-6–induced activation of the STAT3 pathway. Furthermore, the administration of recombinant CTRP4 protein ameliorated disease symptoms. In conclusion, our results indicate that CTRP4, as an endogenous regulator of the IL-6 receptor–signaling pathway, may be a potential therapeutic intervention for Th17-driven autoimmune diseases.

Authors

Lulu Cao, Jinhai Deng, Wei Chen, Minwei He, Ning Zhao, He Huang, Lu Ling, Qi Li, Xiaoxin Zhu, Lu Wang

×

Interferon-α receptor antisense oligonucleotides reduce neuroinflammation and neuropathology in a mouse model of cerebral interferonopathy
Barney Viengkhou, … , Fredrik Kamme, Markus J. Hofer
Barney Viengkhou, … , Fredrik Kamme, Markus J. Hofer
Published February 15, 2024
Citation Information: J Clin Invest. 2024;134(4):e169562. https://doi.org/10.1172/JCI169562.
View: Text | PDF

Interferon-α receptor antisense oligonucleotides reduce neuroinflammation and neuropathology in a mouse model of cerebral interferonopathy

  • Text
  • PDF
Abstract

Chronic and elevated levels of the antiviral cytokine IFN-α in the brain are neurotoxic. This is best observed in patients with genetic cerebral interferonopathies such as Aicardi-Goutières syndrome. Cerebral interferonopathies typically manifest in early childhood and lead to debilitating disease and premature death. There is no cure for these diseases with existing treatments largely aimed at managing symptoms. Thus, an effective therapeutic strategy is urgently needed. Here, we investigated the effect of antisense oligonucleotides targeting the murine IFN-α receptor (Ifnar1 ASOs) in a transgenic mouse model of cerebral interferonopathy. Intracerebroventricular injection of Ifnar1 ASOs into transgenic mice with brain-targeted chronic IFN-α production resulted in a blunted cerebral interferon signature, reduced neuroinflammation, restoration of blood-brain barrier integrity, absence of tissue destruction, and lessened neuronal damage. Remarkably, Ifnar1 ASO treatment was also effective when given after the onset of neuropathological changes, as it reversed such disease-related features. We conclude that ASOs targeting the IFN-α receptor halt and reverse progression of IFN-α–mediated neuroinflammation and neurotoxicity, opening what we believe to be a new and promising approach for the treatment of patients with cerebral interferonopathies.

Authors

Barney Viengkhou, Christine Hong, Curt Mazur, Sagar Damle, Nicholas B. Gallo, Terry C. Fang, Kate Henry, Iain L. Campbell, Fredrik Kamme, Markus J. Hofer

×

Dynamic metabolism of endothelial triglycerides protects against atherosclerosis in mice
Nabil E. Boutagy, … , Carlos Fernandez-Hernando, William C. Sessa
Nabil E. Boutagy, … , Carlos Fernandez-Hernando, William C. Sessa
Published January 4, 2024
Citation Information: J Clin Invest. 2024;134(4):e170453. https://doi.org/10.1172/JCI170453.
View: Text | PDF

Dynamic metabolism of endothelial triglycerides protects against atherosclerosis in mice

  • Text
  • PDF
Abstract

Blood vessels are continually exposed to circulating lipids, and elevation of ApoB-containing lipoproteins causes atherosclerosis. Lipoprotein metabolism is highly regulated by lipolysis, largely at the level of the capillary endothelium lining metabolically active tissues. How large blood vessels, the site of atherosclerotic vascular disease, regulate the flux of fatty acids (FAs) into triglyceride-rich (TG-rich) lipid droplets (LDs) is not known. In this study, we showed that deletion of the enzyme adipose TG lipase (ATGL) in the endothelium led to neutral lipid accumulation in vessels and impaired endothelial-dependent vascular tone and nitric oxide synthesis to promote endothelial dysfunction. Mechanistically, the loss of ATGL led to endoplasmic reticulum stress–induced inflammation in the endothelium. Consistent with this mechanism, deletion of endothelial ATGL markedly increased lesion size in a model of atherosclerosis. Together, these data demonstrate that the dynamics of FA flux through LD affects endothelial cell homeostasis and consequently large vessel function during normal physiology and in a chronic disease state.

Authors

Nabil E. Boutagy, Ana Gamez-Mendez, Joseph W.M. Fowler, Hanming Zhang, Bal K. Chaube, Enric Esplugues, Andrew Kuo, Sungwoon Lee, Daiki Horikami, Jiasheng Zhang, Kathryn M. Citrin, Abhishek K. Singh, Brian G. Coon, Monica Y. Lee, Yajaira Suarez, Carlos Fernandez-Hernando, William C. Sessa

×

68Ga-FAPI PET imaging monitors response to combined TGF-βR inhibition and immunotherapy in metastatic colorectal cancer
Ke Li, … , Shaoli Song, Shuang Tang
Ke Li, … , Shaoli Song, Shuang Tang
Published January 4, 2024
Citation Information: J Clin Invest. 2024;134(4):e170490. https://doi.org/10.1172/JCI170490.
View: Text | PDF | Corrigendum Clinical Research and Public Health

68Ga-FAPI PET imaging monitors response to combined TGF-βR inhibition and immunotherapy in metastatic colorectal cancer

  • Text
  • PDF
Abstract

BACKGROUND Improving and predicting tumor response to immunotherapy remains challenging. Combination therapy with a transforming growth factor-β receptor (TGF-βR) inhibitor that targets cancer-associated fibroblasts (CAFs) is promising for the enhancement of efficacy of immunotherapies. However, the effect of this approach in clinical trials is limited, requiring in vivo methods to better assess tumor responses to combination therapy.METHODS We measured CAFs in vivo using the 68Ga-labeled fibroblast activation protein inhibitor-04 (68Ga-FAPI-04) for PET/CT imaging to guide the combination of TGF-β inhibition and immunotherapy. One hundred thirty-one patients with metastatic colorectal cancer (CRC) underwent 68Ga-FAPI and 18F-fluorodeoxyglucose (18F-FDG) PET/CT imaging. The relationship between uptake of 68Ga-FAPI and tumor immunity was analyzed in patients. Mouse cohorts of metastatic CRC were treated with the TGF-βR inhibitor combined with KN046, which blocks programmed death ligand 1 (PD-L1) and CTLA-4, followed by 68Ga-FAPI and 18F-FDG micro-PET/CT imaging to assess tumor responses.RESULTS Patients with metastatic CRC demonstrated high uptake rates of 68Ga-FAPI, along with suppressive tumor immunity and poor prognosis. The TGF-βR inhibitor enhanced tumor-infiltrating T cells and significantly sensitized metastatic CRC to KN046. 68Ga-FAPI PET/CT imaging accurately monitored the dynamic changes of CAFs and tumor response to combined the TGF-βR inhibitor with immunotherapy.CONCLUSION 68Ga-FAPI PET/CT imaging is powerful in assessing tumor immunity and the response to immunotherapy in metastatic CRC. This study supports future clinical application of 68Ga-FAPI PET/CT to guide precise TGF-β inhibition plus immunotherapy in CRC patients, recommending 68Ga-FAPI and 18F-FDG dual PET/CT for CRC management.TRIAL REGISTRATION CFFSTS Trial, ChiCTR2100053984, Chinese Clinical Trial Registry.FUNDING National Natural Science Foundation of China (82072695, 32270767, 82272035, 81972260).

Authors

Ke Li, Wei Liu, Hang Yu, Jiwei Chen, Wenxuan Tang, Jianpeng Wang, Ming Qi, Yuyun Sun, Xiaoping Xu, Ji Zhang, Xinxiang Li, Weijian Guo, Xiaoling Li, Shaoli Song, Shuang Tang

×

Bik promotes proteasomal degradation to control low-grade inflammation
Yohannes A. Mebratu, … , Scott Randell, Yohannes Tesfaigzi
Yohannes A. Mebratu, … , Scott Randell, Yohannes Tesfaigzi
Published December 19, 2023
Citation Information: J Clin Invest. 2024;134(4):e170594. https://doi.org/10.1172/JCI170594.
View: Text | PDF

Bik promotes proteasomal degradation to control low-grade inflammation

  • Text
  • PDF
Abstract

Although chronic low-grade inflammation does not cause immediate clinical symptoms, over the longer term, it can enhance other insults or age-dependent damage to organ systems and thereby contribute to age-related disorders, such as respiratory disorders, heart disease, metabolic disorders, autoimmunity, and cancer. However, the molecular mechanisms governing low-level inflammation are largely unknown. We discovered that Bcl-2–interacting killer (Bik) deficiency causes low-level inflammation even at baseline and the development of spontaneous emphysema in female but not male mice. Similarly, a single nucleotide polymorphism that reduced Bik levels was associated with increased inflammation and enhanced decline in lung function in humans. Transgenic expression of Bik in the airways of Bik-deficient mice inhibited allergen- or LPS-induced lung inflammation and reversed emphysema in female mice. Bik deficiency increased nuclear but not cytosolic p65 levels because Bik, by modifying the BH4 domain of Bcl-2, interacted with regulatory particle non-ATPase 1 (RPN1) and RPN2 and enhanced proteasomal degradation of nuclear proteins. Bik deficiency increased inflammation primarily in females because Bcl-2 and Bik levels were reduced in lung tissues and airway cells of female compared with male mice. Therefore, controlling low-grade inflammation by modifying the unappreciated role of Bik and Bcl-2 in facilitating proteasomal degradation of nuclear proteins may be crucial in treating chronic age-related diseases.

Authors

Yohannes A. Mebratu, Jane T. Jones, Congjian Liu, Zerihun H. Negasi, Mizanur Rahman, Joselyn Rojas-Quintero, George T. O’Connor, Wei Gao, Josée Dupuis, Michael H. Cho, Augusto A. Litonjua, Scott Randell, Yohannes Tesfaigzi

×

Piezo1 agonist restores meningeal lymphatic vessels, drainage, and brain-CSF perfusion in craniosynostosis and aged mice
Matt J. Matrongolo, … , Young-Kwon Hong, Max A. Tischfield
Matt J. Matrongolo, … , Young-Kwon Hong, Max A. Tischfield
Published November 2, 2023
Citation Information: J Clin Invest. 2024;134(4):e171468. https://doi.org/10.1172/JCI171468.
View: Text | PDF

Piezo1 agonist restores meningeal lymphatic vessels, drainage, and brain-CSF perfusion in craniosynostosis and aged mice

  • Text
  • PDF
Abstract

Skull development coincides with the onset of cerebrospinal fluid (CSF) circulation, brain-CSF perfusion, and meningeal lymphangiogenesis, processes essential for brain waste clearance. How these processes are affected by craniofacial disorders such as craniosynostosis are poorly understood. We report that raised intracranial pressure and diminished CSF flow in craniosynostosis mouse models associate with pathological changes to meningeal lymphatic vessels that affect their sprouting, expansion, and long-term maintenance. We also show that craniosynostosis affects CSF circulatory pathways and perfusion into the brain. Further, craniosynostosis exacerbates amyloid pathology and plaque buildup in Twist1+/–:5xFAD transgenic Alzheimer’s disease models. Treating craniosynostosis mice with Yoda1, a small molecule agonist for Piezo1, reduces intracranial pressure and improves CSF flow, in addition to restoring meningeal lymphangiogenesis, drainage to the deep cervical lymph nodes, and brain-CSF perfusion. Leveraging these findings, we show that Yoda1 treatments in aged mice with reduced CSF flow and turnover improve lymphatic networks, drainage, and brain-CSF perfusion. Our results suggest that CSF provides mechanical force to facilitate meningeal lymphatic growth and maintenance. Additionally, applying Yoda1 agonist in conditions with raised intracranial pressure and/or diminished CSF flow, as seen in craniosynostosis or with ageing, is a possible therapeutic option to help restore meningeal lymphatic networks and brain-CSF perfusion.

Authors

Matt J. Matrongolo, Phillip S. Ang, Junbing Wu, Aditya Jain, Joshua K. Thackray, Akash Reddy, Chi Chang Sung, Gaëtan Barbet, Young-Kwon Hong, Max A. Tischfield

×

p53 suppresses lipid droplet–fueled tumorigenesis through phosphatidylcholine
Xiuduan Xu, … , Peng Li, Peng Jiang
Xiuduan Xu, … , Peng Li, Peng Jiang
Published January 9, 2024
Citation Information: J Clin Invest. 2024;134(4):e171788. https://doi.org/10.1172/JCI171788.
View: Text | PDF

p53 suppresses lipid droplet–fueled tumorigenesis through phosphatidylcholine

  • Text
  • PDF
Abstract

Choline deficiency causes disorders including hepatic abnormalities and is associated with an increased risk of multiple types of cancer. Here, by choline-free diet–associated RNA-Seq analyses, we found that the tumor suppressor p53 drives the Kennedy pathway via PCYT1B to control the growth of lipid droplets (LDs) and their fueling role in tumorigenesis. Mechanistically, through upregulation of PCYT1B, p53 channeled depleted choline stores to phosphatidylcholine (PC) biosynthesis during choline starvation, thus preventing LD coalescence. Cells lacking p53 failed to complete this response to choline depletion, leading to hepatic steatosis and tumorigenesis, and these effects could be reversed by enforcement of PCYT1B expression or restoration of PC abundance. Furthermore, loss of p53 or defects in the Kennedy pathway increased surface localization of hormone-sensitive lipase on LDs to release specific fatty acids that fueled tumor cells in vivo and in vitro. Thus, p53 loss leads to dysregulation of choline metabolism and LD growth and couples perturbed LD homeostasis to tumorigenesis.

Authors

Xiuduan Xu, Jianqin Wang, Li Xu, Peng Li, Peng Jiang

×

Hyperactive mTORC1 in lung mesenchyme induces endothelial cell dysfunction and pulmonary vascular remodeling
Susan M. Lin, … , Jillian F. Evans, Vera P. Krymskaya
Susan M. Lin, … , Jillian F. Evans, Vera P. Krymskaya
Published December 21, 2023
Citation Information: J Clin Invest. 2024;134(4):e172116. https://doi.org/10.1172/JCI172116.
View: Text | PDF

Hyperactive mTORC1 in lung mesenchyme induces endothelial cell dysfunction and pulmonary vascular remodeling

  • Text
  • PDF
Abstract

Lymphangioleiomyomatosis (LAM) is a progressive cystic lung disease caused by tuberous sclerosis complex 1/2 (TSC1/2) gene mutations in pulmonary mesenchymal cells, resulting in activation of the mechanistic target of rapamycin complex 1 (mTORC1). A subset of patients with LAM develop pulmonary vascular remodeling and pulmonary hypertension. Little, however, is known regarding how LAM cells communicate with endothelial cells (ECs) to trigger vascular remodeling. In end-stage LAM lung explants, we identified EC dysfunction characterized by increased EC proliferation and migration, defective angiogenesis, and dysmorphic endothelial tube network formation. To model LAM disease, we used an mTORC1 gain-of-function mouse model with a Tsc2 KO (Tsc2KO) specific to lung mesenchyme (Tbx4LME-Cre Tsc2fl/fl), similar to the mesenchyme-specific genetic alterations seen in human disease. As early as 8 weeks of age, ECs from mice exhibited marked transcriptomic changes despite an absence of morphological changes to the distal lung microvasculature. In contrast, 1-year-old Tbx4LME-Cre Tsc2fl/fl mice spontaneously developed pulmonary vascular remodeling with increased medial thickness. Single-cell RNA-Seq of 1-year-old mouse lung cells identified paracrine ligands originating from Tsc2KO mesenchyme, which can signal through receptors in arterial ECs. These ECs had transcriptionally altered genes including those in pathways associated with blood vessel remodeling. The proposed pathophysiologic mesenchymal ligand–EC receptor crosstalk highlights the importance of an altered mesenchymal cell/EC axis in LAM and other hyperactive mTORC1–driven diseases. Since ECs in patients with LAM and in Tbx4LME-Cre Tsc2fl/fl mice did not harbor TSC2 mutations, our study demonstrates that constitutively active mTORC1 lung mesenchymal cells orchestrated dysfunctional EC responses that contributed to pulmonary vascular remodeling.

Authors

Susan M. Lin, Ryan Rue, Alexander R. Mukhitov, Akansha Goel, Maria C. Basil, Kseniya Obraztsova, Apoorva Babu, Slaven Crnkovic, Owen A. Ledwell, Laura T. Ferguson, Joseph D. Planer, Ana N. Nottingham, Kanth Swaroop Vanka, Carly J. Smith, Edward Cantu III, Grazyna Kwapiszewska, Edward E. Morrisey, Jillian F. Evans, Vera P. Krymskaya

×

The interaction of Synapsin 2a and Synaptogyrin-3 regulates fear extinction in mice
Xi-Ya Shen, … , Ling-Qiang Zhu, Dan Liu
Xi-Ya Shen, … , Ling-Qiang Zhu, Dan Liu
Published January 4, 2024
Citation Information: J Clin Invest. 2024;134(4):e172802. https://doi.org/10.1172/JCI172802.
View: Text | PDF

The interaction of Synapsin 2a and Synaptogyrin-3 regulates fear extinction in mice

  • Text
  • PDF
Abstract

The mechanisms behind a lack of efficient fear extinction in some individuals are unclear. Here, by employing a principal components analysis–based approach, we differentiated the mice into extinction-resistant and susceptible groups. We determined that elevated synapsin 2a (Syn2a) in the infralimbic cortex (IL) to basolateral amygdala (BLA) circuit disrupted presynaptic orchestration, leading to an excitatory/inhibitory imbalance in the BLA region and causing extinction resistance. Overexpression or silencing of Syn2a levels in IL neurons replicated or alleviated behavioral, electrophysiological, and biochemical phenotypes in resistant mice. We further identified that the proline-rich domain H in the C-terminus of Syn2a was indispensable for the interaction with synaptogyrin-3 (Syngr3) and demonstrated that disrupting this interaction restored extinction impairments. Molecular docking revealed that ritonavir, an FDA-approved HIV drug, could disrupt Syn2a-Syngr3 binding and rescue fear extinction behavior in Syn2a-elevated mice. In summary, the aberrant elevation of Syn2a expression and its interaction with Syngr3 at the presynaptic site were crucial in fear extinction resistance, suggesting a potential therapeutic avenue for related disorders.

Authors

Xi-Ya Shen, Juan Zhang, He-Zhou Huang, Shao-Dan Li, Ling Zhou, Shi-Ping Wu, Cheng Tang, Xian Huang, Zhi-Qiang Liu, Zi-Yuan Guo, Xiang Li, Heng-Ye Man, You-Ming Lu, Ling-Qiang Zhu, Dan Liu

×

ACSS2 gene variants determine kidney disease risk by controlling de novo lipogenesis in kidney tubules
Dhanunjay Mukhi, … , Kathryn E. Wellen, Katalin Susztak
Dhanunjay Mukhi, … , Kathryn E. Wellen, Katalin Susztak
Published December 5, 2023
Citation Information: J Clin Invest. 2024;134(4):e172963. https://doi.org/10.1172/JCI172963.
View: Text | PDF

ACSS2 gene variants determine kidney disease risk by controlling de novo lipogenesis in kidney tubules

  • Text
  • PDF
Abstract

Worldwide, over 800 million people are affected by kidney disease, yet its pathogenesis remains elusive, hindering the development of novel therapeutics. In this study, we used kidney-specific expression of quantitative traits and single-nucleus open chromatin analysis to show that genetic variants linked to kidney dysfunction on chromosome 20 target the acyl-CoA synthetase short-chain family 2 (ACSS2). By generating ACSS2-KO mice, we demonstrated their protection from kidney fibrosis in multiple disease models. Our analysis of primary tubular cells revealed that ACSS2 regulated de novo lipogenesis (DNL), causing NADPH depletion and increasing ROS levels, ultimately leading to NLRP3-dependent pyroptosis. Additionally, we discovered that pharmacological inhibition or genetic ablation of fatty acid synthase safeguarded kidney cells against profibrotic gene expression and prevented kidney disease in mice. Lipid accumulation and the expression of genes related to DNL were elevated in the kidneys of patients with fibrosis. Our findings pinpoint ACSS2 as a critical kidney disease gene and reveal the role of DNL in kidney disease.

Authors

Dhanunjay Mukhi, Lingzhi Li, Hongbo Liu, Tomohito Doke, Lakshmi P. Kolligundla, Eunji Ha, Konstantin Kloetzer, Amin Abedini, Sarmistha Mukherjee, Junnan Wu, Poonam Dhillon, Hailong Hu, Dongyin Guan, Katsuhiko Funai, Kahealani Uehara, Paul M. Titchenell, Joseph A. Baur, Kathryn E. Wellen, Katalin Susztak

×

An engineered immunomodulatory IgG1 Fc suppresses autoimmune inflammation through pathways shared with i.v. immunoglobulin
Sunny L. Sneed, … , Pamela B. Conley, Robert M. Anthony
Sunny L. Sneed, … , Pamela B. Conley, Robert M. Anthony
Published February 15, 2024
Citation Information: J Clin Invest. 2024;134(4):e172980. https://doi.org/10.1172/JCI172980.
View: Text | PDF

An engineered immunomodulatory IgG1 Fc suppresses autoimmune inflammation through pathways shared with i.v. immunoglobulin

  • Text
  • PDF
Abstract

Immunoglobulin G (IgG) antibodies in the form of high-dose intravenous immunoglobulin (IVIG) exert immunomodulatory activity and are used in this capacity to treat inflammatory and autoimmune diseases. Reductionist approaches have revealed that terminal sialylation of the single asparagine-linked (N-linked) glycan at position 297 of the IgG1 Fc bestows antiinflammatory activity, which can be recapitulated by introduction of an F241A point mutation in the IgG1 Fc (FcF241A). Here, we examined the antiinflammatory activity of CHO-K1 cell–produced FcF241A in vivo in models of autoimmune inflammation and found it to be independent of sialylation. Intriguingly, sialylation markedly improved the half-life and bioavailability of FcF241A via impaired interaction with the asialoglycoprotein receptor ASGPR. Further, FcF241A suppressed inflammation through the same molecular pathways as IVIG and sialylated IgG1 Fc and required the C-type lectin SIGN-R1 in vivo. This contrasted with FcAbdeg (efgartigimod), an engineered IgG1 Fc with enhanced neonatal Fc receptor (FcRn) binding, which reduced total serum IgG concentrations, independent of SIGN-R1. When coadministered, FcF241A and FcAbdeg exhibited combinatorial antiinflammatory activity. Together, these results demonstrated that the antiinflammatory activity of FcF241A requires SIGN-R1, similarly to that of high-dose IVIG and sialylated IgG1, and can be used in combination with other antiinflammatory therapeutics that rely on divergent pathways, including FcAbdeg.

Authors

Sunny L. Sneed, Brian B. Reese, Ana F.S. Laureano, Sneha Ratnapriya, Isabella Fraschilla, Kate L. Jeffrey, Greg P. Coffey, Pamela B. Conley, Robert M. Anthony

×

Aster-B–dependent estradiol synthesis protects female mice from diet-induced obesity
Xu Xiao, … , John W.R. Schwabe, Peter Tontonoz
Xu Xiao, … , John W.R. Schwabe, Peter Tontonoz
Published January 4, 2024
Citation Information: J Clin Invest. 2024;134(4):e173002. https://doi.org/10.1172/JCI173002.
View: Text | PDF

Aster-B–dependent estradiol synthesis protects female mice from diet-induced obesity

  • Text
  • PDF
Abstract

Aster proteins mediate the nonvesicular transport of cholesterol from the plasma membrane (PM) to the endoplasmic reticulum (ER). However, the importance of nonvesicular sterol movement for physiology and pathophysiology in various tissues is incompletely understood. Here we show that loss of Aster-B leads to diet-induced obesity in female but not in male mice, and that this sex difference is abolished by ovariectomy. We further demonstrate that Aster-B deficiency impairs nonvesicular cholesterol transport from the PM to the ER in ovaries in vivo, leading to hypogonadism and reduced estradiol synthesis. Female Aster-B–deficient mice exhibit reduced locomotor activity and energy expenditure, consistent with established effects of estrogens on systemic metabolism. Administration of exogenous estradiol ameliorates the diet-induced obesity phenotype of Aster-B–deficient female mice. These findings highlight the key role of Aster-B–dependent nonvesicular cholesterol transport in regulating estradiol production and protecting females from obesity.

Authors

Xu Xiao, John P. Kennelly, An-Chieh Feng, Lijing Cheng, Beatriz Romartinez-Alonso, Alexander Bedard, Yajing Gao, Liujuan Cui, Stephen G. Young, John W.R. Schwabe, Peter Tontonoz

×

KCTD1/KCTD15 complexes control ectodermal and neural crest cell functions, and their impairment causes aplasia cutis
Jackelyn R. Raymundo, … , Luigi Vitagliano, Alexander G. Marneros
Jackelyn R. Raymundo, … , Luigi Vitagliano, Alexander G. Marneros
Published December 19, 2023
Citation Information: J Clin Invest. 2024;134(4):e174138. https://doi.org/10.1172/JCI174138.
View: Text | PDF

KCTD1/KCTD15 complexes control ectodermal and neural crest cell functions, and their impairment causes aplasia cutis

  • Text
  • PDF
Abstract

Aplasia cutis congenita (ACC) is a congenital epidermal defect of the midline scalp and has been proposed to be due to a primary keratinocyte abnormality. Why it forms mainly at this anatomic site has remained a long-standing enigma. KCTD1 mutations cause ACC, ectodermal abnormalities, and kidney fibrosis, whereas KCTD15 mutations cause ACC and cardiac outflow tract abnormalities. Here, we found that KCTD1 and KCTD15 can form multimeric complexes and can compensate for each other’s loss and that disease mutations are dominant negative, resulting in lack of KCTD1/KCTD15 function. We demonstrated that KCTD15 is critical for cardiac outflow tract development, whereas KCTD1 regulates distal nephron function. Combined inactivation of KCTD1/KCTD15 in keratinocytes resulted in abnormal skin appendages but not in ACC. Instead, KCTD1/KCTD15 inactivation in neural crest cells resulted in ACC linked to midline skull defects, demonstrating that ACC is not caused by a primary defect in keratinocytes but is a secondary consequence of impaired cranial neural crest cells, giving rise to midline cranial suture cells that express keratinocyte-promoting growth factors. Our findings explain the clinical observations in patients with KCTD1 versus KCTD15 mutations, establish KCTD1/KCTD15 complexes as critical regulators of ectodermal and neural crest cell functions, and define ACC as a neurocristopathy.

Authors

Jackelyn R. Raymundo, Hui Zhang, Giovanni Smaldone, Wenjuan Zhu, Kathleen E. Daly, Benjamin J. Glennon, Giovanni Pecoraro, Marco Salvatore, William A. Devine, Cecilia W. Lo, Luigi Vitagliano, Alexander G. Marneros

×

A metabolic signature for NADSYN1-dependent congenital NAD deficiency disorder
Justin O. Szot, … , Kayleigh Bozon, Sally L. Dunwoodie
Justin O. Szot, … , Kayleigh Bozon, Sally L. Dunwoodie
Published February 15, 2024
Citation Information: J Clin Invest. 2024;134(4):e174824. https://doi.org/10.1172/JCI174824.
View: Text | PDF

A metabolic signature for NADSYN1-dependent congenital NAD deficiency disorder

  • Text
  • PDF
Abstract

Nicotinamide adenine dinucleotide (NAD) is essential for embryonic development. To date, biallelic loss-of-function variants in 3 genes encoding nonredundant enzymes of the NAD de novo synthesis pathway — KYNU, HAAO, and NADSYN1 — have been identified in humans with congenital malformations defined as congenital NAD deficiency disorder (CNDD). Here, we identified 13 further individuals with biallelic NADSYN1 variants predicted to be damaging, and phenotypes ranging from multiple severe malformations to the complete absence of malformation. Enzymatic assessment of variant deleteriousness in vitro revealed protein domain–specific perturbation, complemented by protein structure modeling in silico. We reproduced NADSYN1-dependent CNDD in mice and assessed various maternal NAD precursor supplementation strategies to prevent adverse pregnancy outcomes. While for Nadsyn1+/– mothers, any B3 vitamer was suitable to raise NAD, preventing embryo loss and malformation, Nadsyn1–/– mothers required supplementation with amidated NAD precursors (nicotinamide or nicotinamide mononucleotide) bypassing their metabolic block. The circulatory NAD metabolome in mice and humans before and after NAD precursor supplementation revealed a consistent metabolic signature with utility for patient identification. Our data collectively improve clinical diagnostics of NADSYN1-dependent CNDD, provide guidance for the therapeutic prevention of CNDD, and suggest an ongoing need to maintain NAD levels via amidated NAD precursor supplementation after birth.

Authors

Justin O. Szot, Hartmut Cuny, Ella M.M.A. Martin, Delicia Z. Sheng, Kavitha Iyer, Stephanie Portelli, Vivien Nguyen, Jessica M. Gereis, Dimuthu Alankarage, David Chitayat, Karen Chong, Ingrid M. Wentzensen, Catherine Vincent-Delormé, Alban Lermine, Emma Burkitt-Wright, Weizhen Ji, Lauren Jeffries, Lynn S. Pais, Tiong Y. Tan, James Pitt, Cheryl A. Wise, Helen Wright, Israel D. Andrews, Brianna Pruniski, Theresa A. Grebe, Nicole Corsten-Janssen, Katelijne Bouman, Cathryn Poulton, Supraja Prakash, Boris Keren, Natasha J. Brown, Matthew F. Hunter, Oliver Heath, Saquib A. Lakhani, John H. McDermott, David B. Ascher, Gavin Chapman, Kayleigh Bozon, Sally L. Dunwoodie

×

S1PR1 inhibition induces proapoptotic signaling in T cells and limits humoral responses within lymph nodes
Dhaval Dixit, … , Jordan E. Axelrad, Susan R. Schwab
Dhaval Dixit, … , Jordan E. Axelrad, Susan R. Schwab
Published January 9, 2024
Citation Information: J Clin Invest. 2024;134(4):e174984. https://doi.org/10.1172/JCI174984.
View: Text | PDF

S1PR1 inhibition induces proapoptotic signaling in T cells and limits humoral responses within lymph nodes

  • Text
  • PDF
Abstract

Effective immunity requires a large, diverse naive T cell repertoire circulating among lymphoid organs in search of antigen. Sphingosine 1-phosphate (S1P) and its receptor S1PR1 contribute by both directing T cell migration and supporting T cell survival. Here, we addressed how S1P enables T cell survival and the implications for patients treated with S1PR1 antagonists. We found that S1PR1 limited apoptosis by maintaining the appropriate balance of BCL2 family members via restraint of JNK activity. Interestingly, the same residues of S1PR1 that enable receptor internalization were required to prevent this proapoptotic cascade. Findings in mice were recapitulated in ulcerative colitis patients treated with the S1PR1 antagonist ozanimod, and the loss of naive T cells limited B cell responses. Our findings highlighted an effect of S1PR1 antagonists on the ability to mount immune responses within lymph nodes, beyond their effect on lymph node egress, and suggested both limitations and additional uses of this important class of drugs.

Authors

Dhaval Dixit, Victoria M. Hallisey, Ethan Y.S. Zhu, Martyna Okuniewska, Ken Cadwell, Jerry E. Chipuk, Jordan E. Axelrad, Susan R. Schwab

×

Proteogenomic analysis reveals cytoplasmic sequestration of RUNX1 by the acute myeloid leukemia–initiating CBFB::MYH11 oncofusion protein
Ryan B. Day, … , Christopher A. Miller, Timothy J. Ley
Ryan B. Day, … , Christopher A. Miller, Timothy J. Ley
Published December 7, 2023
Citation Information: J Clin Invest. 2024;134(4):e176311. https://doi.org/10.1172/JCI176311.
View: Text | PDF

Proteogenomic analysis reveals cytoplasmic sequestration of RUNX1 by the acute myeloid leukemia–initiating CBFB::MYH11 oncofusion protein

  • Text
  • PDF
Abstract

Several canonical translocations produce oncofusion genes that can initiate acute myeloid leukemia (AML). Although each translocation is associated with unique features, the mechanisms responsible remain unclear. While proteins interacting with each oncofusion are known to be relevant for how they act, these interactions have not yet been systematically defined. To address this issue in an unbiased fashion, we fused a promiscuous biotin ligase (TurboID) in-frame with 3 favorable-risk AML oncofusion cDNAs (PML::RARA, RUNX1::RUNX1T1, and CBFB::MYH11) and identified their interacting proteins in primary murine hematopoietic cells. The PML::RARA- and RUNX1::RUNX1T1-TurboID fusion proteins labeled common and unique nuclear repressor complexes, implying their nuclear localization. However, CBFB::MYH11-TurboID–interacting proteins were largely cytoplasmic, probably because of an interaction of the MYH11 domain with several cytoplasmic myosin-related proteins. Using a variety of methods, we showed that the CBFB domain of CBFB::MYH11 sequesters RUNX1 in cytoplasmic aggregates; these findings were confirmed in primary human AML cells. Paradoxically, CBFB::MYH11 expression was associated with increased RUNX1/2 expression, suggesting the presence of a sensor for reduced functional RUNX1 protein, and a feedback loop that may attempt to compensate by increasing RUNX1/2 transcription. These findings may have broad implications for AML pathogenesis.

Authors

Ryan B. Day, Julia A. Hickman, Ziheng Xu, Casey D.S. Katerndahl, Francesca Ferraro, Sai Mukund Ramakrishnan, Petra Erdmann-Gilmore, Robert W. Sprung, Yiling Mi, R. Reid Townsend, Christopher A. Miller, Timothy J. Ley

×
Corrigendum
PHGDH is required for germinal center formation and is a therapeutic target in MYC-driven lymphoma
Annalisa D’Avola, … , Karen H. Vousden, John C. Riches
Annalisa D’Avola, … , Karen H. Vousden, John C. Riches
Published February 15, 2024
Citation Information: J Clin Invest. 2024;134(4):e179917. https://doi.org/10.1172/JCI179917.
View: Text | PDF | Amended Article

PHGDH is required for germinal center formation and is a therapeutic target in MYC-driven lymphoma

  • Text
  • PDF
Abstract

Authors

Annalisa D’Avola, Nathalie Legrave, Mylène Tajan, Probir Chakravarty, Ryan L. Shearer, Hamish W. King, Katarina Kluckova, Eric C. Cheung, Andrew J. Clear, Arief S. Gunawan, Lingling Zhang, Louisa K. James, James I. MacRae, John G. Gribben, Dinis P. Calado, Karen H. Vousden, John C. Riches

×
Advertisement

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

Sign up for email alerts