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 November 1, 2019 Previous issue | Next issue

  • Volume 129, Issue 11
Go to section:
  • Conversations with Giants in Medicine
  • Obituary
  • Viewpoints
  • Commentaries
  • Research Articles
  • Retraction

On the cover: ANGPTL4 destabilizes the retinal vasculature in diabetic macular edema

Anti-VEGF therapy, the standard of care for patients with diabetic macular edema (DME), does not substantially improve vision in many treated patients. In this issue of the JCI, Sodhi et al. explore the role of another protein, ANGPTL4, in driving vascular leakage in DME. Their work revealed that ANGPTL4 and VEGF work in concert to destabilize the retina’s vasculature. ANGPTL4’s binding to neuropilin 1 and 2 on endothelial cells disrupts vascular barriers by activating RhoA/ROCK signaling. Treating diabetic mice with a soluble neuropilin 1 fragment blocked ANGPTL1-induced vascular leakage, supporting a potential therapeutic avenue for interfering in ANGPTL4-mediated mechanisms in DME. This issue’s cover depicts the pathological leakage of the retinal vasculature in a patient with diabetic eye disease. Image courtesy of Wilmer Photography; modified by Isabella and Adriana Sodhi.

Conversations with Giants in Medicine
A conversation with Elaine Fuchs
Ushma S. Neill
Ushma S. Neill
Published November 1, 2019
Citation Information: J Clin Invest. 2019;129(11):4551-4552. https://doi.org/10.1172/JCI133402.
View: Text | PDF

A conversation with Elaine Fuchs

  • Text
  • PDF
Abstract

Authors

Ushma S. Neill

×
Obituary
A tribute to Gerald Weissmann (1930–2019)
Steven B. Abramson, … , Mark R. Philips, Charles N. Serhan
Steven B. Abramson, … , Mark R. Philips, Charles N. Serhan
Published October 7, 2019
Citation Information: J Clin Invest. 2019;129(11):4553-4555. https://doi.org/10.1172/JCI133490.
View: Text | PDF

A tribute to Gerald Weissmann (1930–2019)

  • Text
  • PDF
Abstract

Authors

Steven B. Abramson, Paul J. Anderson, Jill P. Buyon, Bruce N. Cronstein, Thoru Pederson, Mark R. Philips, Charles N. Serhan

×
Viewpoints
Trials and tribulations of diabetes: a patient’s perspective
Mary M. Meyer
Mary M. Meyer
Published September 23, 2019
Citation Information: J Clin Invest. 2019;129(11):4556-4557. https://doi.org/10.1172/JCI132144.
View: Text | PDF

Trials and tribulations of diabetes: a patient’s perspective

  • Text
  • PDF
Abstract

Authors

Mary M. Meyer

×

Obtaining prescription drugs in America: it’s no bargain
Arthur L. Caplan
Arthur L. Caplan
Published September 23, 2019
Citation Information: J Clin Invest. 2019;129(11):4558-4559. https://doi.org/10.1172/JCI132977.
View: Text | PDF

Obtaining prescription drugs in America: it’s no bargain

  • Text
  • PDF
Abstract

Authors

Arthur L. Caplan

×

Success in science: what we can learn from women artists
Lona Mody, … , Joel D. Howell, Sanjay Saint
Lona Mody, … , Joel D. Howell, Sanjay Saint
Published September 30, 2019
Citation Information: J Clin Invest. 2019;129(11):4560-4562. https://doi.org/10.1172/JCI130899.
View: Text | PDF

Success in science: what we can learn from women artists

  • Text
  • PDF
Abstract

Authors

Lona Mody, Joel D. Howell, Sanjay Saint

×
Commentaries
Hungry for your alanine: when liver depends on muscle proteolysis
Theresia Sarabhai, Michael Roden
Theresia Sarabhai, Michael Roden
Published September 23, 2019
Citation Information: J Clin Invest. 2019;129(11):4563-4566. https://doi.org/10.1172/JCI131931.
View: Text | PDF

Hungry for your alanine: when liver depends on muscle proteolysis

  • Text
  • PDF
Abstract

Fasting requires complex endocrine and metabolic interorgan crosstalk, which involves shifting from glucose to fatty acid oxidation, derived from adipose tissue lipolysis, in order to preserve glucose for the brain. The glucose-alanine (Cahill) cycle is critical for regenerating glucose. In this issue of JCI, Petersen et al. report on their use of an innovative stable isotope tracer method to show that skeletal muscle–derived alanine becomes rate controlling for hepatic mitochondrial oxidation and, in turn, for glucose production during prolonged fasting. These results provide new insight into skeletal muscle–liver metabolic crosstalk during the fed-to-fasting transition in humans.

Authors

Theresia Sarabhai, Michael Roden

×

Time to change weight gain recommendations for pregnant women with obesity
Sarah S. Comstock
Sarah S. Comstock
Published September 23, 2019
Citation Information: J Clin Invest. 2019;129(11):4567-4569. https://doi.org/10.1172/JCI131932.
View: Text | PDF

Time to change weight gain recommendations for pregnant women with obesity

  • Text
  • PDF
Abstract

Obesity during pregnancy is a major health problem in the United States. In this issue of the JCI, Most et al. fill an important gap in our understanding of energy homeostasis in pregnancy. The researchers measured energy intake, energy expenditure, and body composition in obese pregnant women. They demonstrated that energy intake need not increase in order for obese women to gain the recommended amounts of weight during pregnancy. Additionally, all of the gestational weight gain scenarios (inadequate, recommended, or excess) resulted in similar maternal and fetal perinatal outcomes. This evidence should guide new recommendations on this important topic.

Authors

Sarah S. Comstock

×

Better living through peptide-conjugated chemistry: next-generation antisense oligonucleotides
Elizabeth M. McNally, Brian D. Leverson
Elizabeth M. McNally, Brian D. Leverson
Published September 30, 2019
Citation Information: J Clin Invest. 2019;129(11):4570-4571. https://doi.org/10.1172/JCI131933.
View: Text | PDF

Better living through peptide-conjugated chemistry: next-generation antisense oligonucleotides

  • Text
  • PDF
Abstract

Two different antisense oligonucleotide–based (ASO-based) therapies are currently in clinical use to treat neuromuscular diseases. This success, for Duchenne muscular dystrophy and spinal muscular atrophy, offers hope not only for additional neuromuscular diseases, but also for other disorders that could benefit from RNA-targeted therapies. A major limitation for more widespread application of ASOs relates to relatively poor tissue penetration. In this issue of the JCI, Klein et al. showed that conjugating an ASO with an arginine-rich cell-penetrating peptide, Pip6a, enhanced delivery, resulting in corrective outcome for a mouse model of myotonic dystrophy. Linking ASOs to cell-penetrating peptides, or even other moieties, is an approach currently under development with treatment potential to expand to other disorders.

Authors

Elizabeth M. McNally, Brian D. Leverson

×

Simplified steps to heterologous prime-boost HIV vaccine development?
Nelson L. Michael
Nelson L. Michael
Published September 30, 2019
Citation Information: J Clin Invest. 2019;129(11):4572-4573. https://doi.org/10.1172/JCI132440.
View: Text | PDF

Simplified steps to heterologous prime-boost HIV vaccine development?

  • Text
  • PDF
Abstract

The RV 144 HIV vaccine efficacy study showed a reduction in HIV-1 infection risk in Thai volunteers who received two priming vaccinations of vCP1521 ALVAC (attenuated recombinant canarypox virus expressing HIV group–specific antigen, polymerase, and envelope genes) followed by two additional ALVAC vaccinations and coadministration of purified bivalent gp120 proteins (AIDSVAX B/E). In this issue of the JCI, Rouphael et al. build on these results by substituting a DNA plasmid cocktail expressing HIV-1 subtype C group–specific antigen, polymerase, and envelope antigen genes (DNA-HIV-PT123) for ALVAC in a phase 1b safety and immunogenicity study. The results indicate that the vaccine regimen is safe, elicits promising cross-subtype humoral and cellular responses, and opens up potentially simplified approaches to HIV-1 vaccine development.

Authors

Nelson L. Michael

×

DNA damage response protects against progressive kidney disease
Bruce A. Molitoris
Bruce A. Molitoris
Published October 7, 2019
Citation Information: J Clin Invest. 2019;129(11):4574-4575. https://doi.org/10.1172/JCI131171.
View: Text | PDF

DNA damage response protects against progressive kidney disease

  • Text
  • PDF
Abstract

The pathophysiology of cellular injury and repair has been extensively studied in acute kidney injury (AKI) for more than 70 years. Although a great deal of knowledge has been generated, a debate over the importance of repairing damaged cells versus replacing them by proliferation remains. In this issue of the JCI, Kishi et al. demonstrate that following kidney epithelial cell injury, DNA repair, rather than cell proliferation, plays the central role in recovery and longevity by minimizing apoptosis, G2/M cell-cycle arrest, and subsequent fibrosis. This has important therapeutic implications and highlights the need for more sensitive techniques to evaluate functional, structural, and molecular recovery following injury.

Authors

Bruce A. Molitoris

×

An unbiased approach de-livers unexpected insight into torsin biology
Sarah M. Prophet, Christian Schlieker
Sarah M. Prophet, Christian Schlieker
Published October 7, 2019
Citation Information: J Clin Invest. 2019;129(11):4576-4579. https://doi.org/10.1172/JCI132442.
View: Text | PDF

An unbiased approach de-livers unexpected insight into torsin biology

  • Text
  • PDF
Abstract

Mutations affecting the integrity of the essential torsin ATPase/cofactor system have been identified in a steadily increasing number of congenital disorders. Since most of these mutations affect brain function, much of the research has focused on deciphering disease etiology in the brain. However, torsin is expressed in a wide variety of nonneural tissues and is strictly conserved across species, including the lowest metazoans, suggesting that it plays roles extending beyond neurons. In this issue of the JCI, Shin et al. explored torsin function in the mammalian liver. The group reports major defects in hepatic lipid metabolism when the torsin system is compromised in mice. Remarkably, conditional deletion of either torsinA or its cofactor, lamina-associated polypeptide 1 (LAP1), resulted in fatty liver disease and steatohepatitis, likely from a secretion defect of VLDLs. This study considerably expands our understanding of torsin biology, while providing defined opportunities for future investigations of torsin function and dysfunction in human pathologies.

Authors

Sarah M. Prophet, Christian Schlieker

×

Striking a balance in an antibody network: A roadmap for HIV-1 vaccines
Tysheena P. Charles, Cynthia A. Derdeyn
Tysheena P. Charles, Cynthia A. Derdeyn
Published October 7, 2019
Citation Information: J Clin Invest. 2019;129(11):4580-4582. https://doi.org/10.1172/JCI132535.
View: Text | PDF

Striking a balance in an antibody network: A roadmap for HIV-1 vaccines

  • Text
  • PDF
Abstract

With almost 2 million new HIV-1 infections in 2018, a highly effective vaccine is imperative. Vaccine-elicited HIV-1 antibodies contribute to protection through multiple nonneutralizing activities, but the exact mechanisms remain unknown. In this issue of the JCI, Neidich and associates sought to determine how antibodies contributed to reducing the risk of HIV-1 acquisition in a phase IIb preventative vaccine efficacy trial, HVTN 505. Their studies revealed that antibody-dependent cellular phagocytosis (ADCP) and FcγRIIa binding were strongly associated with reduced HIV-1 risk; however, HIV-1 envelope–specific IgG3, IgA; and host FcγRIIa genotype also influenced risk. This study highlights the intricate interactions between antibodies and innate immune functions in humans.

Authors

Tysheena P. Charles, Cynthia A. Derdeyn

×

Orai1: CRACing the Th17 response in AKI
Sanjeev Noel
Sanjeev Noel
Published October 14, 2019
Citation Information: J Clin Invest. 2019;129(11):4583-4586. https://doi.org/10.1172/JCI131935.
View: Text | PDF

Orai1: CRACing the Th17 response in AKI

  • Text
  • PDF
Abstract

A strong Th17 inflammatory response aggravates ischemia reperfusion–induced (IR-induced) acute kidney injury (AKI), tissue fibrosis, and AKI-to–chronic kidney disease (CKD) progression. However, the underlying mechanisms of sustained Th17 activation following AKI and during AKI-to-CKD progression are unclear. In this issue of the JCI, Mehrotra et al. present compelling evidence that the store-operated calcium (Ca2+) channel Orai1 sustains Th17-driven inflammatory response after AKI and drives the AKI-to-CKD transition. Orai1 blockade significantly protected renal function from IR, attenuated high-salt–induced AKI-to-CKD progression in rats, and decreased Th17 response in rat and human T cells. Therapeutic targeting of Orai1 can potentially reduce AKI, AKI-to-CKD progression, and other Th17-driven diseases.

Authors

Sanjeev Noel

×

Regulating heart repair with cardiac-specific T lymphocytes
Ziad Mallat
Ziad Mallat
Published October 14, 2019
Citation Information: J Clin Invest. 2019;129(11):4587-4589. https://doi.org/10.1172/JCI132441.
View: Text | PDF

Regulating heart repair with cardiac-specific T lymphocytes

  • Text
  • PDF
Abstract

Cardiac tissue necrosis secondary to coronary artery occlusion is one of the most common and deadly sterile injuries in developed countries. In this issue of the JCI, Rieckmann et al. identified and characterized antigen-specific CD4+ T helper (Th) cells that developed in the context of myocardial infarction (MI) in mice. They showed that myosin heavy chain α (MYHCA) is a dominant cardiac autoantigen and that T cells with T cell receptor (TCR) specificity to MYHCA acquired a Treg phenotype when adoptively transferred into infarcted mice, which mediated a cardioprotective healing response. Thus, Rieckmann et al. showed that an acute ischemic insult to the heart, which induces sterile inflammation, promoted, rather than limited, protective T cell autoimmunity. Notably, strategies that support an antigen-specific Treg response may limit the immune-inflammatory response and promote cardiac repair after acute MI.

Authors

Ziad Mallat

×

Hitting the bullseye with a nonlethal payload: resistance in CD123-positive malignancies
Lukasz P. Gondek
Lukasz P. Gondek
Published October 14, 2019
Citation Information: J Clin Invest. 2019;129(11):4590-4592. https://doi.org/10.1172/JCI132443.
View: Text | PDF

Hitting the bullseye with a nonlethal payload: resistance in CD123-positive malignancies

  • Text
  • PDF
Abstract

The interleukin 3 receptor (CD123) is a transmembrane protein that is absent or hardly expressed on normal hematopoietic stem cells, but highly expressed on the surface of cancer cells in several hematologic malignancies. In this issue of the JCI, Togami et al. investigated the mechanism of resistance to the recently approved anti-CD123 agent tagraxofusp, which consists of interleukin 3 fused to a truncated diphtheria toxin (DT) molecule. The authors demonstrated that loss of the intracellular target for DT, diphthamide, a conservative modification of histidine 715 in eukaryotic elongation factor 2, resulted in tagraxofusp resistance. Specifically, hypermethylation of the DPH1 gene, encoding a key enzyme in diphthamide synthesis, resulted in diphthamide loss. Notably, treatment with a DNA hypomethylating agent restored DPH1 expression and resensitized cells to tagraxofusp. The recognition of this resistance mechanism may have important clinical implications and lead to the development of more effective multiagent therapies.

Authors

Lukasz P. Gondek

×
Research Articles
Angiopoietin-like 4 binds neuropilins and cooperates with VEGF to induce diabetic macular edema
Akrit Sodhi, … , Daoyuan Lu, Silvia Montaner
Akrit Sodhi, … , Daoyuan Lu, Silvia Montaner
Published September 23, 2019
Citation Information: J Clin Invest. 2019;129(11):4593-4608. https://doi.org/10.1172/JCI120879.
View: Text | PDF

Angiopoietin-like 4 binds neuropilins and cooperates with VEGF to induce diabetic macular edema

  • Text
  • PDF
Abstract

The majority of patients with diabetic macular edema (DME), the most common cause of vision loss in working-age Americans, do not respond adequately to current therapies targeting VEGFA. Here, we show that expression of angiopoietin-like 4 (ANGPTL4), a HIF-1–regulated gene product, is increased in the eyes of diabetic mice and patients with DME. We observed that ANGPTL4 and VEGF act synergistically to destabilize the retinal vascular barrier. Interestingly, while ANGPTL4 modestly enhanced tyrosine phosphorylation of VEGF receptor 2, promotion of vascular permeability by ANGPTL4 was independent of this receptor. Instead, we found that ANGPTL4 binds directly to neuropilin 1 (NRP1) and NRP2 on endothelial cells (ECs), leading to rapid activation of the RhoA/ROCK signaling pathway and breakdown of EC-EC junctions. Treatment with a soluble fragment of NRP1 (sNRP1) prevented ANGPTL4 from binding to NRP1 and blocked ANGPTL4-induced activation of RhoA as well as EC permeability in vitro and retinal vascular leakage in diabetic animals in vivo. In addition, sNRP1 reduced the stimulation of EC permeability by aqueous fluid from patients with DME. Collectively, these data identify the ANGPTL4/NRP/RhoA pathway as a therapeutic target for the treatment of DME.

Authors

Akrit Sodhi, Tao Ma, Deepak Menon, Monika Deshpande, Kathleen Jee, Aumreetam Dinabandhu, Jordan Vancel, Daoyuan Lu, Silvia Montaner

×

Stromal integrin α11 regulates PDGFRβ signaling and promotes breast cancer progression
Irina Primac, … , Donald Gullberg, Agnès Noel
Irina Primac, … , Donald Gullberg, Agnès Noel
Published July 9, 2019
Citation Information: J Clin Invest. 2019;129(11):4609-4628. https://doi.org/10.1172/JCI125890.
View: Text | PDF

Stromal integrin α11 regulates PDGFRβ signaling and promotes breast cancer progression

  • Text
  • PDF
Abstract

Cancer-associated fibroblasts (CAFs) are key actors in modulating the progression of many solid tumors, such as breast cancer (BC). Herein, we identify an integrin α11/PDGFRβ–positive CAF subset displaying tumor-promoting features in BC. In the preclinical MMTV-PyMT mouse model, integrin α11 deficiency led to a drastic reduction of tumor progression and metastasis. A clear association between integrin α11 and PDGFRβ was found at both transcriptional and histological levels in BC specimens. High stromal integrin α11/PDGFRβ expression was associated with high grades and poorer clinical outcome in human BC patients. Functional assays using 5 CAF subpopulations (1 murine, 4 human) revealed that integrin α11 promotes CAF invasion and CAF-induced tumor cell invasion upon PDGF-BB stimulation. Mechanistically, the proinvasive activity of integrin α11 relies on its ability to interact with PDGFRβ in a ligand-dependent manner and to promote its downstream JNK activation, leading to the production of tenascin C, a proinvasive matricellular protein. Pharmacological inhibition of PDGFRβ and JNK impaired tumor cell invasion induced by integrin α11+ CAFs. Collectively, our study uncovers an integrin α11+ subset of protumoral CAFs that exploits the PDGFRβ/JNK signaling axis to promote tumor invasiveness in BC.

Authors

Irina Primac, Erik Maquoi, Silvia Blacher, Ritva Heljasvaara, Jan Van Deun, Hilde Y.H. Smeland, Annalisa Canale, Thomas Louis, Linda Stuhr, Nor Eddine Sounni, Didier Cataldo, Taina Pihlajaniemi, Christel Pequeux, Olivier De Wever, Donald Gullberg, Agnès Noel

×

HIV-1 in lymph nodes is maintained by cellular proliferation during antiretroviral therapy
William R. McManus, … , John M. Coffin, Mary F. Kearney
William R. McManus, … , John M. Coffin, Mary F. Kearney
Published July 30, 2019
Citation Information: J Clin Invest. 2019;129(11):4629-4642. https://doi.org/10.1172/JCI126714.
View: Text | PDF

HIV-1 in lymph nodes is maintained by cellular proliferation during antiretroviral therapy

  • Text
  • PDF
Abstract

To investigate the possibility that HIV-1 replication in lymph nodes sustains the reservoir during antiretroviral therapy (ART), we looked for evidence of viral replication in 5 donors after up to 13 years of viral suppression. We characterized proviral populations in lymph nodes and peripheral blood before and during ART, evaluated the levels of viral RNA expression in single lymph node and blood cells, and characterized the proviral integration sites in paired lymph node and blood samples. Proviruses with identical sequences, identical integration sites, and similar levels of RNA expression were found in lymph nodes and blood samples collected during ART, and no single sequence with significant divergence from the pretherapy population was detected in either blood or lymph nodes. These findings show that all detectable persistent HIV-1 infection is consistent with maintenance in lymph nodes by clonal proliferation of cells infected before ART and not by ongoing viral replication during ART.

Authors

William R. McManus, Michael J. Bale, Jonathan Spindler, Ann Wiegand, Andrew Musick, Sean C. Patro, Michele D. Sobolewski, Victoria K. Musick, Elizabeth M. Anderson, Joshua C. Cyktor, Elias K. Halvas, Wei Shao, Daria Wells, Xiaolin Wu, Brandon F. Keele, Jeffrey M. Milush, Rebecca Hoh, John W. Mellors, Stephen H. Hughes, Steven G. Deeks, John M. Coffin, Mary F. Kearney

×

Peritoneal GATA6+ macrophages function as a portal for Staphylococcus aureus dissemination
Selina K. Jorch, … , Michael J. Hickey, Paul Kubes
Selina K. Jorch, … , Michael J. Hickey, Paul Kubes
Published September 23, 2019
Citation Information: J Clin Invest. 2019;129(11):4643-4656. https://doi.org/10.1172/JCI127286.
View: Text | PDF

Peritoneal GATA6+ macrophages function as a portal for Staphylococcus aureus dissemination

  • Text
  • PDF
Abstract

Essentially all Staphylococcus aureus (S. aureus) bacteria that gain access to the circulation are plucked out of the bloodstream by the intravascular macrophages of the liver — the Kupffer cells. It is also thought that these bacteria are disseminated via the bloodstream to other organs. Our data show that S. aureus inside Kupffer cells grew and escaped across the mesothelium into the peritoneal cavity and immediately infected GATA-binding factor 6–positive (GATA6+) peritoneal cavity macrophages. These macrophages provided a haven for S. aureus, thereby delaying the neutrophilic response in the peritoneum by 48 hours and allowing dissemination to various peritoneal and retroperitoneal organs including the kidneys. In mice deficient in GATA6+ peritoneal macrophages, neutrophils infiltrated more robustly and reduced S. aureus dissemination. Antibiotics administered i.v. did not prevent dissemination into the peritoneum or to the kidneys, whereas peritoneal administration of vancomycin (particularly liposomal vancomycin with optimized intracellular penetrance capacity) reduced kidney infection and mortality, even when administered 24 hours after infection. These data indicate that GATA6+ macrophages within the peritoneal cavity are a conduit of dissemination for i.v. S. aureus, and changing the route of antibiotic delivery could provide a more effective treatment for patients with peritonitis-associated bacterial sepsis.

Authors

Selina K. Jorch, Bas G.J. Surewaard, Mokarram Hossain, Moritz Peiseler, Carsten Deppermann, Jennifer Deng, Ania Bogoslowski, Fardau van der Wal, Abdelwahab Omri, Michael J. Hickey, Paul Kubes

×

Recombinant annexin A6 promotes membrane repair and protects against muscle injury
Alexis R. Demonbreun, … , David Y. Barefield, Elizabeth M. McNally
Alexis R. Demonbreun, … , David Y. Barefield, Elizabeth M. McNally
Published September 23, 2019
Citation Information: J Clin Invest. 2019;129(11):4657-4670. https://doi.org/10.1172/JCI128840.
View: Text | PDF

Recombinant annexin A6 promotes membrane repair and protects against muscle injury

  • Text
  • PDF
Abstract

Membrane repair is essential to cell survival. In skeletal muscle, injury often associates with plasma membrane disruption. Additionally, muscular dystrophy is linked to mutations in genes that produce fragile membranes or reduce membrane repair. Methods to enhance repair and reduce susceptibility to injury could benefit muscle in both acute and chronic injury settings. Annexins are a family of membrane-associated Ca2+-binding proteins implicated in repair, and annexin A6 was previously identified as a genetic modifier of muscle injury and disease. Annexin A6 forms the repair cap over the site of membrane disruption. To elucidate how annexins facilitate repair, we visualized annexin cap formation during injury. We found that annexin cap size positively correlated with increasing Ca2+ concentrations. We also found that annexin overexpression promoted external blebs enriched in Ca2+ and correlated with a reduction of intracellular Ca2+ at the injury site. Annexin A6 overexpression reduced membrane injury, consistent with enhanced repair. Treatment with recombinant annexin A6 protected against acute muscle injury in vitro and in vivo. Moreover, administration of recombinant annexin A6 in a model of muscular dystrophy reduced serum creatinine kinase, a biomarker of disease. These data identify annexins as mediators of membrane-associated Ca2+ release during membrane repair and annexin A6 as a therapeutic target to enhance membrane repair capacity.

Authors

Alexis R. Demonbreun, Katherine S. Fallon, Claire C. Oosterbaan, Elena Bogdanovic, James L. Warner, Jordan J. Sell, Patrick G. Page, Mattia Quattrocelli, David Y. Barefield, Elizabeth M. McNally

×

Regulation of hepatic mitochondrial oxidation by glucose-alanine cycling during starvation in humans
Kitt Falk Petersen, … , Gary W. Cline, Gerald I. Shulman
Kitt Falk Petersen, … , Gary W. Cline, Gerald I. Shulman
Published September 23, 2019
Citation Information: J Clin Invest. 2019;129(11):4671-4675. https://doi.org/10.1172/JCI129913.
View: Text | PDF Concise Communication

Regulation of hepatic mitochondrial oxidation by glucose-alanine cycling during starvation in humans

  • Text
  • PDF
Abstract

In order to determine whether the glucose-alanine cycle regulates rates of hepatic mitochondrial oxidation in humans, we applied positional isotopomer NMR tracer analysis (PINTA) to assess rates of hepatic mitochondrial oxidation and pyruvate carboxylase flux in healthy volunteers following both an overnight (12 hours) and a 60-hour fast. Following the 60-hour fast, rates of endogenous glucose production and mitochondrial oxidation decreased, whereas rates of hepatic pyruvate carboxylase flux remained unchanged. These reductions were associated with reduced rates of alanine turnover, assessed by [3-13C]alanine, in a subgroup of participants under similar fasting conditions. In order to determine whether this reduction in alanine turnover was responsible for the reduced rates of hepatic mitochondrial oxidation, we infused unlabeled alanine into another subgroup of 60-hour fasted subjects to increase rates of alanine turnover, similar to what was measured after a 12-hour fast, and found that this perturbation increased rates of hepatic mitochondrial oxidation. Taken together, these studies demonstrate that 60 hours of starvation induce marked reductions in rates of hepatic mitochondrial oxidation, which in turn can be attributed to reduced rates of glucose-alanine cycling, and reveal a heretofore undescribed role for glucose-alanine in the regulation of hepatic mitochondrial oxidation in humans.

Authors

Kitt Falk Petersen, Sylvie Dufour, Gary W. Cline, Gerald I. Shulman

×

Elastase 3B mutation links to familial pancreatitis with diabetes and pancreatic adenocarcinoma
Paul C. Moore, … , Mark S. Anderson, Scott A. Oakes
Paul C. Moore, … , Mark S. Anderson, Scott A. Oakes
Published August 1, 2019
Citation Information: J Clin Invest. 2019;129(11):4676-4681. https://doi.org/10.1172/JCI129961.
View: Text | PDF Concise Communication

Elastase 3B mutation links to familial pancreatitis with diabetes and pancreatic adenocarcinoma

  • Text
  • PDF
Abstract

Although improvements in genetic analysis have greatly enhanced our understanding of the mechanisms behind pancreatitis, it continues to afflict many families for whom the hereditary factors remain unknown. Recent evaluation of a patient with a strong family history of pancreatitis prompted us to reexamine a large kindred originally reported over 50 years ago with an autosomal-dominant inheritance pattern of chronic pancreatitis, diabetes, and pancreatic adenocarcinoma. Whole-exome sequencing analysis identified a rare missense mutation in the gene encoding pancreas-specific protease elastase 3B (CELA3B) that cosegregates with disease. Studies of the mutant protein in vitro, in cell lines, and in CRISPR-Cas9–engineered mice indicate that this mutation causes translational upregulation of CELA3B, which, upon secretion and activation by trypsin, leads to uncontrolled proteolysis and recurrent pancreatitis. Although lesions in several other pancreatic proteases have been previously linked to hereditary pancreatitis, to our knowledge, this is the first known instance of a mutation in CELA3B and a defect in translational control contributing to this disease.

Authors

Paul C. Moore, Jessica T. Cortez, Chester E. Chamberlain, Diana Alba, Amy C. Berger, Zoe Quandt, Alice Chan, Mickie H. Cheng, Jhoanne L. Bautista, Justin Peng, Michael S. German, Mark S. Anderson, Scott A. Oakes

×

Evidence-based recommendations for energy intake in pregnant women with obesity
Jasper Most, … , Eric Ravussin, Leanne M. Redman
Jasper Most, … , Eric Ravussin, Leanne M. Redman
Published August 1, 2019
Citation Information: J Clin Invest. 2019;129(11):4682-4690. https://doi.org/10.1172/JCI130341.
View: Text | PDF Clinical Research and Public Health

Evidence-based recommendations for energy intake in pregnant women with obesity

  • Text
  • PDF
Abstract

BACKGROUND In women with obesity, excess gestational weight gain (≥270 g/week) occurs in 2 out of 3 pregnancies and contributes to metabolic impairments in both mother and baby. To improve obstetrical care, objectively assessed information on energy balance is urgently needed. The objective of this study was to characterize determinants of gestational weight gain in women with obesity.METHODS This was a prospective, observational study of pregnant women with obesity. The primary outcome was energy intake calculated by the energy intake-balance method. Energy expenditure was measured by doubly labeled water and whole-room indirect calorimetry and body composition as a 3-compartment model by air displacement plethysmography and isotope dilution in early (13–16 weeks) and late (35–37 weeks) pregnancy.RESULTS In pregnant women with obesity (n = 54), recommended weight gain (n = 8, 15%) during the second and third trimesters was achieved when energy intake was 125 ± 52 kcal/d less than energy expenditure. In contrast, women with excess weight gain (67%) consumed 186 ± 29 kcal/d more than they expended (P < 0.001). Energy balance affected maternal adiposity (recommended: –2.5 ± 0.8 kg fat mass; excess: +2.2 ± 0.5; inadequate: –4.5 ± 0.5; P < 0.001) but not fetal growth. Weight gain was not related to demographics, activity, metabolic biomarkers, or diet quality. We estimated that energy intake requirements for recommended weight gain during the second and third trimesters were not increased as compared with energy requirements early in pregnancy (34 ± 53 kcal/d, P = 0.83).CONCLUSION We here provide what we believe are the first evidence-based recommendations for energy intake in pregnant women with obesity. Contrary to current recommendations, energy intake should not exceed energy expenditure.TRIAL REGISTRATION ClinicalTrials.gov, NCT01954342.FUNDING This study was funded by the National Institutes of Health (R01DK099175) and the Clinical Research Cores at Pennington Biomedical Research Center (U54GM104940 and P30DK072476).

Authors

Jasper Most, Marshall St Amant, Daniel S. Hsia, Abby D. Altazan, Diana M. Thomas, L. Anne Gilmore, Porsha M. Vallo, Robbie A. Beyl, Eric Ravussin, Leanne M. Redman

×

IFN-γ drives inflammatory bowel disease pathogenesis through VE-cadherin–directed vascular barrier disruption
Victoria Langer, … , Nathalie Britzen-Laurent, Michael Stürzl
Victoria Langer, … , Nathalie Britzen-Laurent, Michael Stürzl
Published September 30, 2019
Citation Information: J Clin Invest. 2019;129(11):4691-4707. https://doi.org/10.1172/JCI124884.
View: Text | PDF

IFN-γ drives inflammatory bowel disease pathogenesis through VE-cadherin–directed vascular barrier disruption

  • Text
  • PDF
Abstract

Inflammatory bowel disease (IBD) is a chronic inflammatory disorder with rising incidence. Diseased tissues are heavily vascularized. Surprisingly, the pathogenic impact of the vasculature in IBD and the underlying regulatory mechanisms remain largely unknown. IFN-γ is a major cytokine in IBD pathogenesis, but in the context of the disease, it is almost exclusively its immune-modulatory and epithelial cell–directed functions that have been considered. Recent studies by our group demonstrated that IFN-γ also exerts potent effects on blood vessels. Based on these considerations, we analyzed the vessel-directed pathogenic functions of IFN-γ and found that it drives IBD pathogenesis through vascular barrier disruption. Specifically, we show that inhibition of the IFN-γ response in vessels by endothelial-specific knockout of IFN-γ receptor 2 ameliorates experimentally induced colitis in mice. IFN-γ acts pathogenic by causing a breakdown of the vascular barrier through disruption of the adherens junction protein VE-cadherin. Notably, intestinal vascular barrier dysfunction was also confirmed in human IBD patients, supporting the clinical relevance of our findings. Treatment with imatinib restored VE-cadherin/adherens junctions, inhibited vascular permeability, and significantly reduced colonic inflammation in experimental colitis. Our findings inaugurate the pathogenic impact of IFN-γ–mediated intestinal vessel activation in IBD and open new avenues for vascular-directed treatment of this disease.

Authors

Victoria Langer, Eugenia Vivi, Daniela Regensburger, Thomas H. Winkler, Maximilian J. Waldner, Timo Rath, Benjamin Schmid, Lisa Skottke, Somin Lee, Noo Li Jeon, Thomas Wohlfahrt, Viktoria Kramer, Philipp Tripal, Michael Schumann, Stephan Kersting, Claudia Handtrack, Carol I. Geppert, Karina Suchowski, Ralf H. Adams, Christoph Becker, Andreas Ramming, Elisabeth Naschberger, Nathalie Britzen-Laurent, Michael Stürzl

×

Hotspot SF3B1 mutations induce metabolic reprogramming and vulnerability to serine deprivation
W. Brian Dalton, … , Josh Lauring, Ben Ho Park
W. Brian Dalton, … , Josh Lauring, Ben Ho Park
Published August 8, 2019
Citation Information: J Clin Invest. 2019;129(11):4708-4723. https://doi.org/10.1172/JCI125022.
View: Text | PDF

Hotspot SF3B1 mutations induce metabolic reprogramming and vulnerability to serine deprivation

  • Text
  • PDF
Abstract

Cancer-associated mutations in the spliceosome gene SF3B1 create a neomorphic protein that produces aberrant mRNA splicing in hundreds of genes, but the ensuing biologic and therapeutic consequences of this missplicing are not well understood. Here we have provided evidence that aberrant splicing by mutant SF3B1 altered the transcriptome, proteome, and metabolome of human cells, leading to missplicing-associated downregulation of metabolic genes, decreased mitochondrial respiration, and suppression of the serine synthesis pathway. We also found that mutant SF3B1 induces vulnerability to deprivation of the nonessential amino acid serine, which was mediated by missplicing-associated downregulation of the serine synthesis pathway enzyme PHGDH. This vulnerability was manifest both in vitro and in vivo, as dietary restriction of serine and glycine in mice was able to inhibit the growth of SF3B1MUT xenografts. These findings describe a role for SF3B1 mutations in altered energy metabolism, and they offer a new therapeutic strategy against SF3B1MUT cancers.

Authors

W. Brian Dalton, Eric Helmenstine, Noel Walsh, Lukasz P. Gondek, Dhanashree S. Kelkar, Abigail Read, Rachael Natrajan, Eric S. Christenson, Barbara Roman, Samarjit Das, Liang Zhao, Robert D. Leone, Daniel Shinn, Taylor Groginski, Anil K. Madugundu, Arun Patil, Daniel J. Zabransky, Arielle Medford, Justin Lee, Alex J. Cole, Marc Rosen, Maya Thakar, Alexander Ambinder, Joshua Donaldson, Amy E. DeZern, Karen Cravero, David Chu, Rafael Madero-Marroquin, Akhilesh Pandey, Paula J. Hurley, Josh Lauring, Ben Ho Park

×

FOXN1 compound heterozygous mutations cause selective thymic hypoplasia in humans
Qiumei Du, … , M. Teresa de la Morena, Nicolai S.C. van Oers
Qiumei Du, … , M. Teresa de la Morena, Nicolai S.C. van Oers
Published September 30, 2019
Citation Information: J Clin Invest. 2019;129(11):4724-4738. https://doi.org/10.1172/JCI127565.
View: Text | PDF

FOXN1 compound heterozygous mutations cause selective thymic hypoplasia in humans

  • Text
  • PDF
Abstract

We report on 2 patients with compound heterozygous mutations in forkhead box N1 (FOXN1), a transcription factor essential for thymic epithelial cell (TEC) differentiation. TECs are critical for T cell development. Both patients had a presentation consistent with T–/loB+NK+ SCID, with normal hair and nails, distinct from the classic nude/SCID phenotype in individuals with autosomal-recessive FOXN1 mutations. To understand the basis of this phenotype and the effects of the mutations on FOXN1, we generated mice using CRISPR-Cas9 technology to genocopy mutations in 1 of the patients. The mice with the Foxn1 compound heterozygous mutations had thymic hypoplasia, causing a T–B+NK+ SCID phenotype, whereas the hair and nails of these mice were normal. Characterization of the functional changes due to the Foxn1 mutations revealed a 5–amino acid segment at the end of the DNA-binding domain essential for the development of TECs but not keratinocytes. The transcriptional activity of this Foxn1 mutant was partly retained, indicating a region that specifies TEC functions. Analysis of an additional 9 FOXN1 mutations identified in multiple unrelated patients revealed distinct functional consequences contingent on the impact of the mutation on the DNA-binding and transactivation domains of FOXN1.

Authors

Qiumei Du, Larry K. Huynh, Fatma Coskun, Erika Molina, Matthew A. King, Prithvi Raj, Shaheen Khan, Igor Dozmorov, Christine M. Seroogy, Christian A. Wysocki, Grace T. Padron, Tyler R. Yates, M. Louise Markert, M. Teresa de la Morena, Nicolai S.C. van Oers

×

Peptide-conjugated oligonucleotides evoke long-lasting myotonic dystrophy correction in patient-derived cells and mice
Arnaud F. Klein, … , Denis Furling, Matthew J.A. Wood
Arnaud F. Klein, … , Denis Furling, Matthew J.A. Wood
Published September 3, 2019
Citation Information: J Clin Invest. 2019;129(11):4739-4744. https://doi.org/10.1172/JCI128205.
View: Text | PDF Concise Communication

Peptide-conjugated oligonucleotides evoke long-lasting myotonic dystrophy correction in patient-derived cells and mice

  • Text
  • PDF
Abstract

Antisense oligonucleotides (ASOs) targeting pathologic RNAs have shown promising therapeutic corrections for many genetic diseases including myotonic dystrophy (DM1). Thus, ASO strategies for DM1 can abolish the toxic RNA gain-of-function mechanism caused by nucleus-retained mutant DMPK (DM1 protein kinase) transcripts containing CUG expansions (CUGexps). However, systemic use of ASOs for this muscular disease remains challenging due to poor drug distribution to skeletal muscle. To overcome this limitation, we test an arginine-rich Pip6a cell-penetrating peptide and show that Pip6a-conjugated morpholino phosphorodiamidate oligomer (PMO) dramatically enhanced ASO delivery into striated muscles of DM1 mice following systemic administration in comparison with unconjugated PMO and other ASO strategies. Thus, low-dose treatment with Pip6a-PMO-CAG targeting pathologic expansions is sufficient to reverse both splicing defects and myotonia in DM1 mice and normalizes the overall disease transcriptome. Moreover, treated DM1 patient–derived muscle cells showed that Pip6a-PMO-CAG specifically targets mutant CUGexp-DMPK transcripts to abrogate the detrimental sequestration of MBNL1 splicing factor by nuclear RNA foci and consequently MBNL1 functional loss, responsible for splicing defects and muscle dysfunction. Our results demonstrate that Pip6a-PMO-CAG induces long-lasting correction with high efficacy of DM1-associated phenotypes at both molecular and functional levels, and strongly support the use of advanced peptide conjugates for systemic corrective therapy in DM1.

Authors

Arnaud F. Klein, Miguel A. Varela, Ludovic Arandel, Ashling Holland, Naira Naouar, Andrey Arzumanov, David Seoane, Lucile Revillod, Guillaume Bassez, Arnaud Ferry, Dominic Jauvin, Genevieve Gourdon, Jack Puymirat, Michael J. Gait, Denis Furling, Matthew J.A. Wood

×

JMJD3 regulates CD4+ T cell trafficking by targeting actin cytoskeleton regulatory gene Pdlim4
Chuntang Fu, … , Helen Y. Wang, Rong-Fu Wang
Chuntang Fu, … , Helen Y. Wang, Rong-Fu Wang
Published August 8, 2019
Citation Information: J Clin Invest. 2019;129(11):4745-4757. https://doi.org/10.1172/JCI128293.
View: Text | PDF

JMJD3 regulates CD4+ T cell trafficking by targeting actin cytoskeleton regulatory gene Pdlim4

  • Text
  • PDF
Abstract

Histone H3K27 demethylase JMJD3 plays a critical role in gene expression and T cell differentiation. However, the role and mechanisms of JMJD3 in T cell trafficking remain poorly understood. Here, we show that JMJD3 deficiency in CD4+ T cells resulted in an accumulation of T cells in the thymus and reduction of T cell number in the secondary lymphoid organs. We identified PDLIM4 as a significantly downregulated target gene in JMJD3-deficient CD4+ T cells by gene profiling and ChIP-Seq analyses. We further showed that PDLIM4 functioned as an adaptor protein to interact with sphingosine-1 phosphate receptor 1 (S1P1) and filamentous actin (F-actin), thus serving as a key regulator of T cell trafficking. Mechanistically, JMJD3 bound to the promoter and gene-body regions of the Pdlim4 gene and regulated its expression by interacting with zinc finger transcription factor KLF2. Our findings have identified Pdlim4 as a JMJD3 target gene that affects T cell trafficking by cooperating with S1P1 and have provided insights into the molecular mechanisms by which JMJD3 regulates genes involved in T cell trafficking.

Authors

Chuntang Fu, Qingtian Li, Jia Zou, Changsheng Xing, Mei Luo, Bingnan Yin, Junjun Chu, Jiaming Yu, Xin Liu, Helen Y. Wang, Rong-Fu Wang

×

Early adaptive immune activation detected in monozygotic twins with prodromal multiple sclerosis
Eduardo Beltrán, … , Reinhard Hohlfeld, Klaus Dornmair
Eduardo Beltrán, … , Reinhard Hohlfeld, Klaus Dornmair
Published September 30, 2019
Citation Information: J Clin Invest. 2019;129(11):4758-4768. https://doi.org/10.1172/JCI128475.
View: Text | PDF

Early adaptive immune activation detected in monozygotic twins with prodromal multiple sclerosis

  • Text
  • PDF
Abstract

Multiple sclerosis (MS) is a disabling disease of the CNS. Inflammatory features of MS include lymphocyte accumulations in the CNS and cerebrospinal fluid (CSF). The preclinical events leading to established MS are still enigmatic. Here we compared gene expression patterns of CSF cells from MS-discordant monozygotic twin pairs. Six “healthy” co-twins, who carry a maximal familial risk for developing MS, showed subclinical neuroinflammation (SCNI) with small MRI lesions. Four of these subjects had oligoclonal bands (OCBs). By single-cell RNA sequencing of 2752 CSF cells, we identified clonally expanded CD8+ T cells, plasmablasts, and, to a lesser extent, CD4+ T cells not only from MS patients but also from subjects with SCNI. In contrast to nonexpanded T cells, clonally expanded T cells showed characteristics of activated tissue-resident memory T (TRM) cells. The TRM-like phenotype was detectable already in cells from SCNI subjects but more pronounced in cells from patients with definite MS. Expanded plasmablast clones were detected only in MS and SCNI subjects with OCBs. Our data provide evidence for very early concomitant activation of 3 components of the adaptive immune system in MS, with a notable contribution of clonally expanded TRM-like CD8+ cells.

Authors

Eduardo Beltrán, Lisa Ann Gerdes, Julia Hansen, Andrea Flierl-Hecht, Stefan Krebs, Helmut Blum, Birgit Ertl-Wagner, Frederik Barkhof, Tania Kümpfel, Reinhard Hohlfeld, Klaus Dornmair

×

DNA priming and gp120 boosting induces HIV-specific antibodies in a randomized clinical trial
Nadine G. Rouphael, … , Michael C. Keefer, the HVTN 105 Protocol Team and the NIAID HIV Vaccine Trials Network
Nadine G. Rouphael, … , Michael C. Keefer, the HVTN 105 Protocol Team and the NIAID HIV Vaccine Trials Network
Published September 30, 2019
Citation Information: J Clin Invest. 2019;129(11):4769-4785. https://doi.org/10.1172/JCI128699.
View: Text | PDF Clinical Research and Public Health

DNA priming and gp120 boosting induces HIV-specific antibodies in a randomized clinical trial

  • Text
  • PDF
Abstract

BACKGROUND RV144 is the only preventive HIV vaccine regimen demonstrating efficacy in humans. Attempting to build upon RV144 immune responses, we conducted a phase 1, multicenter, randomized, double-blind trial to assess the safety and immunogenicity of regimens substituting the DNA-HIV-PT123 (DNA) vaccine for ALVAC-HIV in different sequences or combinations with AIDSVAX B/E (protein).METHODS One hundred and four HIV-uninfected participants were randomized to 4 treatment groups (T1, T2, T3, and T4) and received intramuscular injections at 0, 1, 3, and 6 months (M): T1 received protein at M0 and M1 and DNA at M3 and M6; T2 received DNA at M0 and M1 and protein at M3 and M6; T3 received DNA at M0, M1, M3, and M6 with protein coadministered at M3 and M6; and T4 received protein and DNA coadministered at each vaccination visit.RESULTS All regimens were well tolerated. Antibodies binding to gp120 and V1V2 scaffold were observed in 95%–100% of participants in T3 and T4, two weeks after final vaccination at high magnitude. While IgG3 responses were highest in T3, a lower IgA/IgG ratio was observed in T4. Binding antibodies persisted at 12 months in 35%–100% of participants. Antibody-dependent cell-mediated cytotoxicity and tier 1 neutralizing-antibody responses had higher response rates for T3 and T4, respectively. CD4+ T cell responses were detectable in all treatment groups (32%–64%) without appreciable CD8+ T cell responses.CONCLUSION The DNA/protein combination regimens induced high-magnitude and long-lasting HIV V1V2–binding antibody responses, and early coadministration of the 2 vaccines led to a more rapid induction of these potentially protective responses.TRIAL REGISTRATION ClinicalTrials.gov NCT02207920.FUNDING National Institute of Allergy and Infectious Diseases (NIAID) grants UM1 AI068614, UM1 AI068635, UM1 AI068618, UM1 AI069511, UM1 AI069470, UM1 AI069534, P30 AI450008, UM1 AI069439, UM1 AI069481, and UM1 AI069496; the National Center for Advancing Translational Sciences, NIH (grant UL1TR001873); and the Bill & Melinda Gates Foundation (grant OPP52845).

Authors

Nadine G. Rouphael, Cecilia Morgan, Shuying S. Li, Ryan Jensen, Brittany Sanchez, Shelly Karuna, Edith Swann, Magdalena E. Sobieszczyk, Ian Frank, Gregory J. Wilson, Hong-Van Tieu, Janine Maenza, Aliza Norwood, James Kobie, Faruk Sinangil, Giuseppe Pantaleo, Song Ding, M. Juliana McElrath, Stephen C. De Rosa, David C. Montefiori, Guido Ferrari, Georgia D. Tomaras, Michael C. Keefer, the HVTN 105 Protocol Team and the NIAID HIV Vaccine Trials Network

×

Plasma deconvolution identifies broadly neutralizing antibodies associated with hepatitis C virus clearance
Valerie J. Kinchen, … , James E. Crowe Jr, Justin R. Bailey
Valerie J. Kinchen, … , James E. Crowe Jr, Justin R. Bailey
Published August 13, 2019
Citation Information: J Clin Invest. 2019;129(11):4786-4796. https://doi.org/10.1172/JCI130720.
View: Text | PDF

Plasma deconvolution identifies broadly neutralizing antibodies associated with hepatitis C virus clearance

  • Text
  • PDF
Abstract

A vaccine for hepatitis C virus (HCV) is urgently needed. Development of broadly neutralizing plasma antibodies during acute infection is associated with HCV clearance, but the viral epitopes of these plasma antibodies are unknown. Identifying these epitopes could define the specificity and function of neutralizing antibodies (NAbs) that should be induced by a vaccine. Here, we present the development and application of a high-throughput method that deconvolutes polyclonal anti-HCV NAbs in plasma, delineating the epitope specificities of anti-HCV NAbs in acute-infection plasma of 44 humans with subsequent clearance or persistence of HCV. Remarkably, we identified multiple broadly neutralizing antibody combinations that were associated with greater plasma neutralizing breadth and with HCV clearance. These studies have the potential to inform new strategies for vaccine development by identifying broadly neutralizing antibody combinations in plasma associated with the natural clearance of HCV, while also providing a high-throughput assay that could identify these responses after vaccination trials.

Authors

Valerie J. Kinchen, Guido Massaccesi, Andrew I. Flyak, Madeleine C. Mankowski, Michelle D. Colbert, William O. Osburn, Stuart C. Ray, Andrea L. Cox, James E. Crowe Jr, Justin R. Bailey

×

Proximal tubule ATR regulates DNA repair to prevent maladaptive renal injury responses
Seiji Kishi, … , Ryuji Morizane, Joseph V. Bonventre
Seiji Kishi, … , Ryuji Morizane, Joseph V. Bonventre
Published October 7, 2019
Citation Information: J Clin Invest. 2019;129(11):4797-4816. https://doi.org/10.1172/JCI122313.
View: Text | PDF

Proximal tubule ATR regulates DNA repair to prevent maladaptive renal injury responses

  • Text
  • PDF
Abstract

Maladaptive proximal tubule (PT) repair has been implicated in kidney fibrosis through induction of cell-cycle arrest at G2/M. We explored the relative importance of the PT DNA damage response (DDR) in kidney fibrosis by genetically inactivating ataxia telangiectasia and Rad3-related (ATR), which is a sensor and upstream initiator of the DDR. In human chronic kidney disease, ATR expression inversely correlates with DNA damage. ATR was upregulated in approximately 70% of Lotus tetragonolobus lectin–positive (LTL+) PT cells in cisplatin-exposed human kidney organoids. Inhibition of ATR resulted in greater PT cell injury in organoids and cultured PT cells. PT-specific Atr-knockout (ATRRPTC–/–) mice exhibited greater kidney function impairment, DNA damage, and fibrosis than did WT mice in response to kidney injury induced by either cisplatin, bilateral ischemia-reperfusion, or unilateral ureteral obstruction. ATRRPTC–/– mice had more cells in the G2/M phase after injury than did WT mice after similar treatments. In conclusion, PT ATR activation is a key component of the DDR, which confers a protective effect mitigating the maladaptive repair and consequent fibrosis that follow kidney injury.

Authors

Seiji Kishi, Craig R. Brooks, Kensei Taguchi, Takaharu Ichimura, Yutaro Mori, Akinwande Akinfolarin, Navin Gupta, Pierre Galichon, Bertha C. Elias, Tomohisa Suzuki, Qian Wang, Leslie Gewin, Ryuji Morizane, Joseph V. Bonventre

×

Age-dependent SMN expression in disease-relevant tissue and implications for SMA treatment
Daniel M. Ramos, … , Thomas O. Crawford, Charlotte J. Sumner
Daniel M. Ramos, … , Thomas O. Crawford, Charlotte J. Sumner
Published October 7, 2019
Citation Information: J Clin Invest. 2019;129(11):4817-4831. https://doi.org/10.1172/JCI124120.
View: Text | PDF Clinical Research and Public Health

Age-dependent SMN expression in disease-relevant tissue and implications for SMA treatment

  • Text
  • PDF
Abstract

BACKGROUND Spinal muscular atrophy (SMA) is caused by deficient expression of survival motor neuron (SMN) protein. New SMN-enhancing therapeutics are associated with variable clinical benefits. Limited knowledge of baseline and drug-induced SMN levels in disease-relevant tissues hinders efforts to optimize these treatments.METHODS SMN mRNA and protein levels were quantified in human tissues isolated during expedited autopsies.RESULTS SMN protein expression varied broadly among prenatal control spinal cord samples, but was restricted at relatively low levels in controls and SMA patients after 3 months of life. A 2.3-fold perinatal decrease in median SMN protein levels was not paralleled by comparable changes in SMN mRNA. In tissues isolated from nusinersen-treated SMA patients, antisense oligonucleotide (ASO) concentration and full-length (exon 7 including) SMN2 (SMN2-FL) mRNA level increases were highest in lumbar and thoracic spinal cord. An increased number of cells showed SMN immunolabeling in spinal cord of treated patients, but was not associated with an increase in whole-tissue SMN protein levels.CONCLUSIONS A normally occurring perinatal decrease in whole-tissue SMN protein levels supports efforts to initiate SMN-inducing therapies as soon after birth as possible. Limited ASO distribution to rostral spinal and brain regions in some patients likely limits clinical response of motor units in these regions for those patients. These results have important implications for optimizing treatment of SMA patients and warrant further investigations to enhance bioavailability of intrathecally administered ASOs.FUNDING SMA Foundation, SMART, NIH (R01-NS096770, R01-NS062869), Ionis Pharmaceuticals, and PTC Therapeutics. Biogen provided support for absolute real-time RT-PCR.

Authors

Daniel M. Ramos, Constantin d’Ydewalle, Vijayalakshmi Gabbeta, Amal Dakka, Stephanie K. Klein, Daniel A. Norris, John Matson, Shannon J. Taylor, Phillip G. Zaworski, Thomas W. Prior, Pamela J. Snyder, David Valdivia, Christine L. Hatem, Ian Waters, Nikhil Gupte, Kathryn J. Swoboda, Frank Rigo, C. Frank Bennett, Nikolai Naryshkin, Sergey Paushkin, Thomas O. Crawford, Charlotte J. Sumner

×

Glycan-dependent HIV-specific neutralizing antibodies bind to cells of uninfected individuals
Jana Blazkova, … , Susan Moir, Tae-Wook Chun
Jana Blazkova, … , Susan Moir, Tae-Wook Chun
Published October 7, 2019
Citation Information: J Clin Invest. 2019;129(11):4832-4837. https://doi.org/10.1172/JCI125955.
View: Text | PDF Concise Communication

Glycan-dependent HIV-specific neutralizing antibodies bind to cells of uninfected individuals

  • Text
  • PDF
Abstract

A number of highly potent and broadly neutralizing antibodies (bNAbs) against the human immunodeficiency virus (HIV) have recently been shown to prevent transmission of the virus, suppress viral replication, and delay plasma viral rebound following discontinuation of antiretroviral therapy in animal models and infected humans. However, the degree and extent to which such bNAbs interact with primary lymphocytes have not been fully delineated. Here, we show that certain glycan-dependent bNAbs, such as PGT121 and PGT151, bind to B, activated T, and natural killer (NK) cells of HIV-infected and -uninfected individuals. Binding of these bNAbs, particularly PGT121 and PGT151, to activated CD4+ and CD8+ T cells was mediated by complex-type glycans and was abrogated by enzymatic inhibition of N-linked glycosylation. In addition, a short-term incubation of PGT151 and primary NK cells led to degranulation and cellular death. Our data suggest that the propensity of certain bNAbs to bind uninfected/bystander cells has the potential for unexpected outcomes in passive-transfer studies and underscore the importance of antibody screening against primary lymphocytes.

Authors

Jana Blazkova, Eric W. Refsland, Katherine E. Clarridge, Victoria Shi, J. Shawn Justement, Erin D. Huiting, Kathleen R. Gittens, Xuejun Chen, Stephen D. Schmidt, Cuiping Liu, Nicole Doria-Rose, John R. Mascola, Alonso Heredia, Susan Moir, Tae-Wook Chun

×

Antibody Fc effector functions and IgG3 associate with decreased HIV-1 risk
Scott D. Neidich, … , Peter B. Gilbert, Georgia D. Tomaras
Scott D. Neidich, … , Peter B. Gilbert, Georgia D. Tomaras
Published October 7, 2019
Citation Information: J Clin Invest. 2019;129(11):4838-4849. https://doi.org/10.1172/JCI126391.
View: Text | PDF

Antibody Fc effector functions and IgG3 associate with decreased HIV-1 risk

  • Text
  • PDF
Abstract

HVTN 505 is a preventative vaccine efficacy trial testing DNA followed by recombinant adenovirus serotype 5 (rAd5) in circumcised, Ad5-seronegative men and transgendered persons who have sex with men in the United States. Identified immune correlates of lower HIV-1 risk and a virus sieve analysis revealed that, despite lacking overall efficacy, vaccine-elicited responses exerted pressure on infecting HIV-1 viruses. To interrogate the mechanism of the antibody correlate of HIV-1 risk, we examined antigen-specific antibody recruitment of Fcγ receptors (FcγRs), antibody-dependent cellular phagocytosis (ADCP), and the role of anti-envelope (anti-Env) IgG3. In a prespecified immune correlates analysis, antibody-dependent monocyte phagocytosis and antibody binding to FcγRIIa correlated with decreased HIV-1 risk. Follow-up analyses revealed that anti-Env IgG3 breadth correlated with reduced HIV-1 risk, anti-Env IgA negatively modified infection risk by Fc effector functions, and that vaccine recipients with a specific FcγRIIa single-nucleotide polymorphism locus had a stronger correlation with decreased HIV-1 risk when ADCP, Env-FcγRIIa, and IgG3 binding were high. Additionally, FcγRIIa engagement correlated with decreased viral load setpoint in vaccine recipients who acquired HIV-1. These data support a role for vaccine-elicited anti–HIV-1 Env IgG3, antibody engagement of FcRs, and phagocytosis as potential mechanisms for HIV-1 prevention.

Authors

Scott D. Neidich, Youyi Fong, Shuying S. Li, Daniel E. Geraghty, Brian D. Williamson, William Chad Young, Derrick Goodman, Kelly E. Seaton, Xiaoying Shen, Sheetal Sawant, Lu Zhang, Allan C. deCamp, Bryan S. Blette, Mengshu Shao, Nicole L. Yates, Frederick Feely, Chul-Woo Pyo, Guido Ferrari, HVTN 505 Team, Ian Frank, Shelly T. Karuna, Edith M. Swann, John R. Mascola, Barney S. Graham, Scott M. Hammer, Magdalena E. Sobieszczyk, Lawrence Corey, Holly E. Janes, M. Juliana McElrath, Raphael Gottardo, Peter B. Gilbert, Georgia D. Tomaras

×

cGAS/STING axis mediates a topoisomerase II inhibitor–induced tumor immunogenicity
Zining Wang, … , Liang-ping Xia, Xiaojun Xia
Zining Wang, … , Liang-ping Xia, Xiaojun Xia
Published August 13, 2019
Citation Information: J Clin Invest. 2019;129(11):4850-4862. https://doi.org/10.1172/JCI127471.
View: Text | PDF

cGAS/STING axis mediates a topoisomerase II inhibitor–induced tumor immunogenicity

  • Text
  • PDF
Abstract

Checkpoint blockade antibodies have been approved as immunotherapy for multiple types of cancer, but the response rate and efficacy are still limited. There are few immunogenic cell death–inducing (ICD-inducing) drugs available that can kill cancer cells, enhance tumor immunogenicity, increase in vivo immune infiltration, and thereby boost a tumor response to immunotherapy. So far, the ICD markers have been identified as the few immunostimulating characteristics of dead cells, but whether the presence of such ICD markers on tumor cells translates into enhanced antitumor immunity in vivo is still being investigated. To identify anticancer drugs that could induce tumor cell death and boost T cell response, we performed drug screenings based on both an ICD reporter assay and a T cell activation assay. We showed that teniposide, a DNA topoisomerase II inhibitor, could induce high-mobility group box 1 (HMGB1) release and type I IFN signaling in tumor cells and that teniposide-treated tumor cells could activate antitumor T cell response both in vitro and in vivo. Mechanistically, teniposide induced tumor cell DNA damage and innate immune signaling, including NF-κB activation and stimulator of IFN genes–dependent (STING-dependent) type I IFN signaling, both of which contribute to the activation of dendritic cells and subsequent T cells. Furthermore, teniposide potentiated the antitumor efficacy of anti-PD1 in multiple types of mouse tumor models. Our findings showed that teniposide could trigger tumor immunogenicity and enabled a potential chemoimmunotherapeutic approach to potentiating the therapeutic efficacy of anti-PD1 immunotherapy.

Authors

Zining Wang, Jiemin Chen, Jie Hu, Hongxia Zhang, Feifei Xu, Wenzhuo He, Xiaojuan Wang, Mengyun Li, Wenhua Lu, Gucheng Zeng, Penghui Zhou, Peng Huang, Siyu Chen, Wende Li, Liang-ping Xia, Xiaojun Xia

×

Integrin α5β1 regulates PP2A complex assembly through PDE4D in atherosclerosis
Sanguk Yun, … , David C. Pallas, Martin A. Schwartz
Sanguk Yun, … , David C. Pallas, Martin A. Schwartz
Published August 13, 2019
Citation Information: J Clin Invest. 2019;129(11):4863-4874. https://doi.org/10.1172/JCI127692.
View: Text | PDF

Integrin α5β1 regulates PP2A complex assembly through PDE4D in atherosclerosis

  • Text
  • PDF
Abstract

Fibronectin in the vascular wall promotes inflammatory activation of the endothelium during vascular remodeling and atherosclerosis. These effects are mediated in part by fibronectin binding to integrin α5, which recruits and activates phosphodiesterase 4D5 (PDE4D5) by inducing its dephosphorylation on an inhibitory site, S651. Active PDE then hydrolyzes antiinflammatory cAMP to facilitate inflammatory signaling. To test this model in vivo, we mutated the integrin binding site of PDE4D5 in mice. This mutation reduced endothelial inflammatory activation in atherosclerosis-prone regions of arteries and, in a hyperlipidemia model, reduced atherosclerotic plaque size while increasing markers of plaque stability. We then investigated the mechanism of PDE4D5 activation. Proteomics identified the PP2A regulatory subunit B55α as the factor recruiting PP2A to PDE4D5. The B55α-PP2A complex localized to adhesions and directly dephosphorylated PDE4D5. This interaction also, unexpectedly, stabilized the PP2A-B55α complex. The integrin-regulated, proatherosclerotic transcription factor Yap was also dephosphorylated and activated through this pathway. PDE4D5 therefore mediated matrix-specific regulation of endothelial cell phenotype via an unconventional adapter role, assembling and anchoring a multifunctional PP2A complex that has other targets. We believe these results may have widespread consequences for the control of cell function by integrins.

Authors

Sanguk Yun, Rui Hu, Melanie E. Schwaemmle, Alexander N. Scherer, Zhenwu Zhuang, Anthony J. Koleske, David C. Pallas, Martin A. Schwartz

×

p53-responsive TLR8 SNP enhances human innate immune response to respiratory syncytial virus
Daniel Menendez, … , Steven R. Kleeberger, Michael A. Resnick
Daniel Menendez, … , Steven R. Kleeberger, Michael A. Resnick
Published August 20, 2019
Citation Information: J Clin Invest. 2019;129(11):4875-4884. https://doi.org/10.1172/JCI128626.
View: Text | PDF

p53-responsive TLR8 SNP enhances human innate immune response to respiratory syncytial virus

  • Text
  • PDF
Abstract

The Toll-like receptor 8 (TLR8) has an important role in innate immune responses to RNA viral infections, including respiratory syncytial virus (RSV). We previously reported that TLR8 expression was increased directly by the tumor suppressor and transcription factor p53 via a single nucleotide polymorphism (SNP) (rs3761624) in the TLR8 promoter, thereby placing TLR8 in the p53/immune axis. Because this SNP is in linkage disequilibrium with other SNPs associated with several infectious diseases, we addressed the combined influence of p53 and the SNP on downstream inflammatory signaling in response to a TLR8 cognate ssRNA ligand. Using human primary lymphocytes, p53 induction by chemotherapeutic agents such as ionizing radiation caused SNP-dependent synergistic increases in IL-6 following incubation with an ssRNA ligand, as well as TLR8 RNA and protein expression along with p53 binding at the TLR-p53 SNP site. Because TLR8 is X-linked, the increases were generally reduced in heterozygous females. We found a corresponding association of the p53-responsive allele with RSV disease severity in infants hospitalized with RSV infection. We conclude that p53 can strongly influence TLR8-mediated immune responses and that knowledge of the p53-responsive SNP can inform diagnosis and prognosis of RSV disease and other diseases that might have a TLR8 component, including cancer.

Authors

Daniel Menendez, Joyce Snipe, Jacqui Marzec, Cynthia L. Innes, Fernando P. Polack, Mauricio T. Caballero, Shepherd H. Schurman, Steven R. Kleeberger, Michael A. Resnick

×

Nuclear envelope–localized torsinA-LAP1 complex regulates hepatic VLDL secretion and steatosis
Ji-Yeon Shin, … , Henry N. Ginsberg, Howard J. Worman
Ji-Yeon Shin, … , Henry N. Ginsberg, Howard J. Worman
Published August 13, 2019
Citation Information: J Clin Invest. 2019;129(11):4885-4900. https://doi.org/10.1172/JCI129769.
View: Text | PDF

Nuclear envelope–localized torsinA-LAP1 complex regulates hepatic VLDL secretion and steatosis

  • Text
  • PDF
Abstract

Deciphering novel pathways that regulate liver lipid content has profound implications for understanding the pathophysiology of nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH). Recent evidence suggests that the nuclear envelope is a site of regulation of lipid metabolism, but there is limited appreciation of the responsible mechanisms and molecular components within this organelle. We showed that conditional hepatocyte deletion of the inner nuclear membrane protein lamina-associated polypeptide 1 (LAP1) causes defective VLDL secretion and steatosis, including intranuclear lipid accumulation. LAP1 binds to and activates torsinA, an AAA+ ATPase that resides in the perinuclear space and continuous main ER. Deletion of torsinA from mouse hepatocytes caused even greater reductions in VLDL secretion and profound steatosis. Mice from both of the mutant lines studied developed hepatic steatosis and subsequent steatohepatitis on a regular chow diet in the absence of whole-body insulin resistance or obesity. Our results establish an essential role for the nuclear envelope–localized torsinA-LAP1 complex in hepatic VLDL secretion and suggest that the torsinA pathway participates in the pathophysiology of NAFLD.

Authors

Ji-Yeon Shin, Antonio Hernandez-Ono, Tatyana Fedotova, Cecilia Östlund, Michael J. Lee, Sarah B. Gibeley, Chun-Chi Liang, William T. Dauer, Henry N. Ginsberg, Howard J. Worman

×

AAV8-vectored suprachoroidal gene transfer produces widespread ocular transgene expression
Kun Ding, … , Olivier Danos, Peter A. Campochiaro
Kun Ding, … , Olivier Danos, Peter A. Campochiaro
Published August 13, 2019
Citation Information: J Clin Invest. 2019;129(11):4901-4911. https://doi.org/10.1172/JCI129085.
View: Text | PDF

AAV8-vectored suprachoroidal gene transfer produces widespread ocular transgene expression

  • Text
  • PDF
Abstract

There has been great progress in ocular gene therapy, but delivery of viral vectors to the retinal pigmented epithelium (RPE) and retina can be challenging. Subretinal injection, the preferred route of delivery for most applications, requires a surgical procedure that has risks. Herein we report a novel gene therapy delivery approach, suprachoroidal injection of AAV8 vectors, which is less invasive and could be done in an outpatient setting. Two weeks after suprachoroidal injection of AAV8.GFP in rats, GFP fluorescence covered 18.9% of RPE flat mounts and extended entirely around sagittal and transverse sections in RPE and photoreceptors. After 2 suprachoroidal injections of AAV8.GFP, GFP fluorescence covered 30.5% of RPE flat mounts. Similarly, widespread expression of GFP occurred in nonhuman primate and pig eyes after suprachoroidal injection of AAV8.GFP. Compared with subretinal injection in rats of RGX-314, an AAV8 vector expressing an anti-VEGF Fab, suprachoroidal injection of the same dose of RGX-314 resulted in similar expression of anti-VEGF Fab and similar suppression of VEGF-induced vascular leakage. Suprachoroidal AAV8 vector injection provides a noninvasive outpatient procedure to obtain widespread transgene expression in retina and RPE.

Authors

Kun Ding, Jikui Shen, Zibran Hafiz, Sean F. Hackett, Raquel Lima e Silva, Mahmood Khan, Valeria E. Lorenc, Daiqin Chen, Rishi Chadha, Minie Zhang, Sherri Van Everen, Nicholas Buss, Michele Fiscella, Olivier Danos, Peter A. Campochiaro

×

Lymphatic mimicry in maternal endothelial cells promotes placental spiral artery remodeling
John B. Pawlak, … , Zoltán Jakus, Kathleen M. Caron
John B. Pawlak, … , Zoltán Jakus, Kathleen M. Caron
Published August 15, 2019
Citation Information: J Clin Invest. 2019;129(11):4912-4921. https://doi.org/10.1172/JCI120446.
View: Text | PDF

Lymphatic mimicry in maternal endothelial cells promotes placental spiral artery remodeling

  • Text
  • PDF
Abstract

Molecular heterogeneity of endothelial cells underlies their highly specialized functions during changing physiological conditions within diverse vascular beds. For example, placental spiral arteries (SAs) undergo remarkable remodeling to meet the ever-growing demands of the fetus — a process which is deficient in preeclampsia. The extent to which maternal endothelial cells coordinate with immune cells and pregnancy hormones to promote SA remodeling remains largely unknown. Here we found that remodeled SAs expressed the lymphatic markers PROX1, LYVE1, and VEGFR3, mimicking lymphatic identity. Uterine natural killer (uNK) cells, which are required for SA remodeling and secrete VEGFC, were both sufficient and necessary for VEGFR3 activation in vitro and in mice lacking uNK cells, respectively. Using Flt4Chy/+ mice with kinase inactive VEGFR3 and Vegfcfl/fl Vav1-Cre mice, we demonstrated that SA remodeling required VEGFR3 signaling, and that disrupted maternal VEGFR3 signaling contributed to late-gestation fetal growth restriction. Collectively, we identified a novel instance of lymphatic mimicry by which maternal endothelial cells promote SA remodeling, furthering our understanding of the vascular heterogeneity employed for the mitigation of pregnancy complications such as fetal growth restriction and preeclampsia.

Authors

John B. Pawlak, László Bálint, Lillian Lim, Wanshu Ma, Reema B. Davis, Zoltán Benyó, Michael J. Soares, Guillermo Oliver, Mark L. Kahn, Zoltán Jakus, Kathleen M. Caron

×

Myocardial infarction triggers cardioprotective antigen-specific T helper cell responses
Max Rieckmann, … , Ulrich Hofmann, Gustavo Campos Ramos
Max Rieckmann, … , Ulrich Hofmann, Gustavo Campos Ramos
Published August 13, 2019
Citation Information: J Clin Invest. 2019;129(11):4922-4936. https://doi.org/10.1172/JCI123859.
View: Text | PDF

Myocardial infarction triggers cardioprotective antigen-specific T helper cell responses

  • Text
  • PDF
Abstract

T cell autoreactivity is a hallmark of autoimmune diseases but can also benefit self-maintenance and foster tissue repair. Here, we investigated whether heart-specific T cells exert salutary or detrimental effects in the context of myocardial infarction (MI), the leading cause of death worldwide. After screening more than 150 class II–restricted epitopes, we found that myosin heavy chain α (MYHCA) was a dominant cardiac antigen triggering post-MI CD4+ T cell activation in Balb/c mice. Transferred MYHCA614–629-specific CD4+ T cells (TCR-M cells) selectively accumulated in the myocardium and mediastinal lymph nodes (med-LNs) of infarcted mice, acquired a Treg phenotype with a distinct prohealing gene expression profile, and mediated cardioprotection. Myocardial Tregs were also detected in autopsy samples from patients who had had a MI. Noninvasive PET/CT imaging using a CXCR4 radioligand revealed enlarged med-LNs with increased cellularity in patients with MI. Notably, the med-LN alterations observed in MI patients correlated with the infarct size and cardiac function. Taken together, the results obtained in our study provide evidence that MI context induces prohealing T cell autoimmunity in mice and confirm the existence of an analogous heart/med-LN/T cell axis in patients with MI.

Authors

Max Rieckmann, Murilo Delgobo, Chiara Gaal, Lotte Büchner, Philipp Steinau, Dan Reshef, Cristina Gil-Cruz, Ellis N. ter Horst, Malte Kircher, Theresa Reiter, Katrin G. Heinze, Hans W.M. Niessen, Paul A.J. Krijnen, Anja M. van der Laan, Jan J. Piek, Charlotte Koch, Hans-Jürgen Wester, Constantin Lapa, Wolfgang R. Bauer, Burkhard Ludewig, Nir Friedman, Stefan Frantz, Ulrich Hofmann, Gustavo Campos Ramos

×

Atrial fibrillation risk loci interact to modulate Ca2+-dependent atrial rhythm homeostasis
Brigitte Laforest, … , Christopher R. Weber, Ivan P. Moskowitz
Brigitte Laforest, … , Christopher R. Weber, Ivan P. Moskowitz
Published October 14, 2019
Citation Information: J Clin Invest. 2019;129(11):4937-4950. https://doi.org/10.1172/JCI124231.
View: Text | PDF

Atrial fibrillation risk loci interact to modulate Ca2+-dependent atrial rhythm homeostasis

  • Text
  • PDF
Abstract

Atrial fibrillation (AF), defined by disorganized atrial cardiac rhythm, is the most prevalent cardiac arrhythmia worldwide. Recent genetic studies have highlighted a major heritable component and identified numerous loci associated with AF risk, including the cardiogenic transcription factor genes TBX5, GATA4, and NKX2-5. We report that Tbx5 and Gata4 interact with opposite signs for atrial rhythm controls compared with cardiac development. Using mouse genetics, we found that AF pathophysiology caused by Tbx5 haploinsufficiency, including atrial arrhythmia susceptibility, prolonged action potential duration, and ectopic cardiomyocyte depolarizations, were all rescued by Gata4 haploinsufficiency. In contrast, Nkx2-5 haploinsufficiency showed no combinatorial effect. The molecular basis of the TBX5/GATA4 interaction included normalization of intra-cardiomyocyte calcium flux and expression of calcium channel genes Atp2a2 and Ryr2. Furthermore, GATA4 and TBX5 showed antagonistic interactions on an Ryr2 enhancer. Atrial rhythm instability caused by Tbx5 haploinsufficiency was rescued by a decreased dose of phospholamban, a sarco/endoplasmic reticulum Ca2+-ATPase inhibitor, consistent with a role for decreased sarcoplasmic reticulum calcium flux in Tbx5-dependent AF susceptibility. This work defines a link between Tbx5 dose, sarcoplasmic reticulum calcium flux, and AF propensity. The unexpected interactions between Tbx5 and Gata4 in atrial rhythm control suggest that evaluating specific interactions between genetic risk loci will be necessary for ascertaining personalized risk from genetic association data.

Authors

Brigitte Laforest, Wenli Dai, Leonid Tyan, Sonja Lazarevic, Kaitlyn M. Shen, Margaret Gadek, Michael T. Broman, Christopher R. Weber, Ivan P. Moskowitz

×

Calcium channel Orai1 promotes lymphocyte IL-17 expression and progressive kidney injury
Purvi Mehrotra, … , Javier A. Neyra, David P. Basile
Purvi Mehrotra, … , Javier A. Neyra, David P. Basile
Published August 15, 2019
Citation Information: J Clin Invest. 2019;129(11):4951-4961. https://doi.org/10.1172/JCI126108.
View: Text | PDF | Corrigendum

Calcium channel Orai1 promotes lymphocyte IL-17 expression and progressive kidney injury

  • Text
  • PDF
Abstract

We hypothesized that the store-operated calcium entry (SOCE) channel, Orai1, participates in the activation of Th17 cells and influences renal injury. In rats, following renal ischemia/reperfusion (I/R), there was a rapid and sustained influx of Orai1+ CD4 T cells and IL-17 expression was restricted to Orai1+ cells. When kidney CD4+ cells of post–acute kidney injury (post-AKI) rats were stimulated with angiotensin II and elevated Na+ (10–7 M/170 mM) in vitro, there was an enhanced response in intracellular Ca2+ and IL-17 expression, which was blocked by SOCE inhibitors 2APB, YM58483/BTP2, or AnCoA4. In vivo, YM58483/BTP2 (1 mg/kg) attenuated IL-17+ cell activation, inflammation, and severity of AKI following either I/R or intramuscular glycerol injection. Rats treated with high-salt diet (5–9 weeks after I/R) manifested progressive disease indicated by enhanced inflammation, fibrosis, and impaired renal function. These responses were significantly attenuated by YM58483/BTP2. In peripheral blood of critically ill patients, Orai1+ cells were significantly elevated by approximately 10-fold and Th17 cells were elevated by approximately 4-fold in AKI versus non-AKI patients. Further, in vitro stimulation of CD4+ cells from AKI patients increased IL-17, which was blocked by SOCE inhibitors. These data suggest that Orai1 SOCE is a potential therapeutic target in AKI and CKD progression.

Authors

Purvi Mehrotra, Michael Sturek, Javier A. Neyra, David P. Basile

×

Increased flux through the mevalonate pathway mediates fibrotic repair without injury
Jennifer L. Larson-Casey, … , Veena B. Antony, A. Brent Carter
Jennifer L. Larson-Casey, … , Veena B. Antony, A. Brent Carter
Published October 14, 2019
Citation Information: J Clin Invest. 2019;129(11):4962-4978. https://doi.org/10.1172/JCI127959.
View: Text | PDF

Increased flux through the mevalonate pathway mediates fibrotic repair without injury

  • Text
  • PDF
Abstract

Macrophages are important in mounting an innate immune response to injury as well as in repair of injury. Gene expression of Rho proteins is known to be increased in fibrotic models; however, the role of these proteins in idiopathic pulmonary fibrosis (IPF) is not known. Here, we show that BAL cells from patients with IPF have a profibrotic phenotype secondary to increased activation of the small GTPase Rac1. Rac1 activation requires a posttranslational modification, geranylgeranylation, of the C-terminal cysteine residue. We found that by supplying more substrate for geranylgeranylation, Rac1 activation was substantially increased, resulting in profibrotic polarization by increasing flux through the mevalonate pathway. The increased flux was secondary to greater levels of acetyl-CoA from metabolic reprogramming to β oxidation. The polarization mediated fibrotic repair in the absence of injury by enhancing macrophage/fibroblast signaling. These observations suggest that targeting the mevalonate pathway may abrogate the role of macrophages in dysregulated fibrotic repair.

Authors

Jennifer L. Larson-Casey, Mudit Vaid, Linlin Gu, Chao He, Guo-Qiang Cai, Qiang Ding, Dana Davis, Taylor F. Berryhill, Landon S. Wilson, Stephen Barnes, Jeffrey D. Neighbors, Raymond J. Hohl, Kurt A. Zimmerman, Bradley K. Yoder, Ana Leda F. Longhini, Vidya Sagar Hanumanthu, Ranu Surolia, Veena B. Antony, A. Brent Carter

×

Airway epithelium–shifted mast cell infiltration regulates asthmatic inflammation via IL-33 signaling
Matthew C. Altman, … , Michael C. Peters, Teal S. Hallstrand
Matthew C. Altman, … , Michael C. Peters, Teal S. Hallstrand
Published August 22, 2019
Citation Information: J Clin Invest. 2019;129(11):4979-4991. https://doi.org/10.1172/JCI126402.
View: Text | PDF

Airway epithelium–shifted mast cell infiltration regulates asthmatic inflammation via IL-33 signaling

  • Text
  • PDF
Abstract

Asthma is a heterogeneous syndrome that has been subdivided into physiologic phenotypes and molecular endotypes. The most specific phenotypic manifestation of asthma is indirect airway hyperresponsiveness (AHR), and a prominent molecular endotype is the presence of type 2 inflammation. The underlying basis for type 2 inflammation and its relationship to AHR are incompletely understood. We assessed the expression of type 2 cytokines in the airways of subjects with and without asthma who were extensively characterized for AHR. Using quantitative morphometry of the airway wall, we identified a shift in mast cells from the submucosa to the airway epithelium specifically associated with both type 2 inflammation and indirect AHR. Using ex vivo modeling of primary airway epithelial cells in organotypic coculture with mast cells, we show that epithelial-derived IL-33 uniquely induced type 2 cytokines in mast cells, which regulated the expression of epithelial IL33 in a feed-forward loop. This feed-forward loop was accentuated in epithelial cells derived from subjects with asthma. These results demonstrate that type 2 inflammation and indirect AHR in asthma are related to a shift in mast cell infiltration to the airway epithelium, and that mast cells cooperate with epithelial cells through IL-33 signaling to regulate type 2 inflammation.

Authors

Matthew C. Altman, Ying Lai, James D. Nolin, Sydney Long, Chien-Chang Chen, Adrian M. Piliponsky, William A. Altemeier, Megan Larmore, Charles W. Frevert, Michael S. Mulligan, Steven F. Ziegler, Jason S. Debley, Michael C. Peters, Teal S. Hallstrand

×

Recognition of human gastrointestinal cancer neoantigens by circulating PD-1+ lymphocytes
Alena Gros, … , James C. Yang, Steven A. Rosenberg
Alena Gros, … , James C. Yang, Steven A. Rosenberg
Published October 14, 2019
Citation Information: J Clin Invest. 2019;129(11):4992-5004. https://doi.org/10.1172/JCI127967.
View: Text | PDF

Recognition of human gastrointestinal cancer neoantigens by circulating PD-1+ lymphocytes

  • Text
  • PDF
Abstract

Tumor-resident lymphocytes can mount a response against neoantigens expressed in microsatellite-stable gastrointestinal (GI) cancers, and adoptive transfer of neoantigen-specific lymphocytes has demonstrated antitumor activity in selected patients. However, whether peripheral blood could be used as an alternative minimally invasive source to identify lymphocytes targeting neoantigens in patients with GI cancer with relatively low mutation burden is unclear. We used a personalized high-throughput screening strategy to investigate whether PD-1 expression in peripheral blood could be used to identify CD8+ or CD4+ lymphocytes recognizing neoantigens identified by whole-exome sequencing in 7 patients with GI cancer. We found that neoantigen-specific lymphocytes were preferentially enriched in the CD8+PD-1+/hi or CD4+PD-1+/hi subsets, but not in the corresponding bulk or PD-1– fractions. In 6 of 7 individuals analyzed we identified circulating CD8+ and CD4+ lymphocytes targeting 6 and 4 neoantigens, respectively. Moreover, neoantigen-reactive T cells and a T cell receptor (TCR) isolated from the CD8+PD-1+ subsets recognized autologous tumor, albeit at reduced levels, in 2 patients with available cell lines. These data demonstrate the existence of circulating T cells targeting neoantigens in GI cancer patients and provide an approach to generate enriched populations of personalized neoantigen-specific lymphocytes and isolate TCRs that could be exploited therapeutically to treat cancer.

Authors

Alena Gros, Eric Tran, Maria R. Parkhurst, Sadia Ilyas, Anna Pasetto, Eric M. Groh, Paul F. Robbins, Rami Yossef, Andrea Garcia-Garijo, Carlos A. Fajardo, Todd D. Prickett, Li Jia, Jared J. Gartner, Satyajit Ray, Lien Ngo, John R. Wunderllich, James C. Yang, Steven A. Rosenberg

×

DNA methyltransferase inhibition overcomes diphthamide pathway deficiencies underlying CD123-targeted treatment resistance
Katsuhiro Togami, … , Cory M. Johannessen, Andrew A. Lane
Katsuhiro Togami, … , Cory M. Johannessen, Andrew A. Lane
Published August 22, 2019
Citation Information: J Clin Invest. 2019;129(11):5005-5019. https://doi.org/10.1172/JCI128571.
View: Text | PDF

DNA methyltransferase inhibition overcomes diphthamide pathway deficiencies underlying CD123-targeted treatment resistance

  • Text
  • PDF
Abstract

The interleukin-3 receptor α subunit, CD123, is expressed in many hematologic malignancies including acute myeloid leukemia (AML) and blastic plasmacytoid dendritic cell neoplasm (BPDCN). Tagraxofusp (SL-401) is a CD123-targeted therapy consisting of interleukin-3 fused to a truncated diphtheria toxin payload. Factors influencing response to tagraxofusp other than CD123 expression are largely unknown. We interrogated tagraxofusp resistance in patients and experimental models and found that it was not associated with CD123 loss. Rather, resistant AML and BPDCN cells frequently acquired deficiencies in the diphthamide synthesis pathway, impairing tagraxofusp’s ability to ADP-ribosylate cellular targets. Expression of DPH1, encoding a diphthamide pathway enzyme, was reduced by DNA CpG methylation in resistant cells. Treatment with the DNA methyltransferase inhibitor azacitidine restored DPH1 expression and tagraxofusp sensitivity. We also developed a drug-dependent ADP-ribosylation assay in primary cells that correlated with tagraxofusp activity and may represent an additional novel biomarker. As predicted by these results and our observation that resistance also increased mitochondrial apoptotic priming, we found that the combination of tagraxofusp and azacitidine was effective in patient-derived xenografts treated in vivo. These data have important implications for clinical use of tagraxofusp and led to a phase 1 study combining tagraxofusp and azacitidine in myeloid malignancies.

Authors

Katsuhiro Togami, Timothy Pastika, Jason Stephansky, Mahmoud Ghandi, Amanda L. Christie, Kristen L. Jones, Carl A. Johnson, Ross W. Lindsay, Christopher L. Brooks, Anthony Letai, Jeffrey W. Craig, Olga Pozdnyakova, David M. Weinstock, Joan Montero, Jon C. Aster, Cory M. Johannessen, Andrew A. Lane

×

T cell repertoire remodeling following post-transplant T cell therapy coincides with clinical response
Corey Smith, … , Daniel Chambers, Rajiv Khanna
Corey Smith, … , Daniel Chambers, Rajiv Khanna
Published August 15, 2019
Citation Information: J Clin Invest. 2019;129(11):5020-5032. https://doi.org/10.1172/JCI128323.
View: Text | PDF Clinical Research and Public Health

T cell repertoire remodeling following post-transplant T cell therapy coincides with clinical response

  • Text
  • PDF
Abstract

BACKGROUND Impaired T cell immunity in transplant recipients is associated with infection-related morbidity and mortality. We recently reported the successful use of adoptive T cell therapy (ACT) against drug-resistant/recurrent cytomegalovirus in solid-organ transplant recipients.METHODS In the present study, we used high-throughput T cell receptor Vβ sequencing and T cell functional profiling to delineate the impact of ACT on T cell repertoire remodeling in the context of pretherapy immunity and ACT products.RESULTS These analyses indicated that a clinical response was coincident with significant changes in the T cell receptor Vβ landscape after therapy. This restructuring was associated with the emergence of effector memory T cells in responding patients, while nonresponders displayed dramatic pretherapy T cell expansions with minimal change following ACT. Furthermore, immune reconstitution included both adoptively transferred clonotypes and endogenous clonotypes not detected in the ACT products.CONCLUSION These observations demonstrate that immune control following ACT requires significant repertoire remodeling, which may be impaired in nonresponders because of the preexisting immune environment. Immunological interventions that can modulate this environment may improve clinical outcomes.TRIAL REGISTRATION Australian New Zealand Clinical Trial Registry, ACTRN12613000981729.FUNDING This study was supported by funding from the National Health and Medical Research Council, Australia (APP1132519 and APP1062074).

Authors

Corey Smith, Dillon Corvino, Leone Beagley, Sweera Rehan, Michelle A. Neller, Pauline Crooks, Katherine K. Matthews, Matthew Solomon, Laetitia Le Texier, Scott Campbell, Ross S. Francis, Daniel Chambers, Rajiv Khanna

×

Myo-inositol oxygenase expression profile modulates pathogenic ferroptosis in the renal proximal tubule
Fei Deng, … , Ming Yang, Yashpal S. Kanwar
Fei Deng, … , Ming Yang, Yashpal S. Kanwar
Published August 22, 2019
Citation Information: J Clin Invest. 2019;129(11):5033-5049. https://doi.org/10.1172/JCI129903.
View: Text | PDF

Myo-inositol oxygenase expression profile modulates pathogenic ferroptosis in the renal proximal tubule

  • Text
  • PDF
Abstract

Overexpression of myo-inositol oxygenase (MIOX), a proximal tubular enzyme, exacerbates cellular redox injury in acute kidney injury (AKI). Ferroptosis, a newly coined term associated with lipid hydroperoxidation, plays a critical role in the pathogenesis of AKI. Whether or not MIOX exacerbates tubular damage by accelerating ferroptosis in cisplatin-induced AKI remains elusive. Cisplatin-treated HK-2 cells exhibited notable cell death, which was reduced by ferroptosis inhibitors. Also, alterations in various ferroptosis metabolic sensors, including lipid hydroperoxidation, glutathione peroxidase 4 (GPX4) activity, NADPH and reduced glutathione (GSH) levels, and ferritinophagy, were observed. These perturbations were accentuated by MIOX overexpression, while ameliorated by MIOX knockdown. Likewise, cisplatin-treated CD1 mice exhibited tubular damage and derangement of renal physiological parameters, which were alleviated by ferrostatin-1, a ferroptosis inhibitor. To investigate the relevance of MIOX to ferroptosis, WT mice, MIOX-overexpressing transgenic (MIOX-Tg) mice, and MIOX-KO mice were subjected to cisplatin treatment. In comparison with cisplatin-treated WT mice, cisplatin-treated MIOX-Tg mice had more severe renal pathological changes and perturbations in ferroptosis metabolic sensors, which were minimal in cisplatin-treated MIOX-KO mice. In conclusion, these findings indicate that ferroptosis, an integral process in the pathogenesis of cisplatin-induced AKI, is modulated by the expression profile of MIOX.

Authors

Fei Deng, Isha Sharma, Yingbo Dai, Ming Yang, Yashpal S. Kanwar

×
Retraction
Targeting ceramide synthase 6–dependent metastasis-prone phenotype in lung cancer cells
Motoshi Suzuki, … , Mamoru Kyogashima, Takashi Takahashi
Motoshi Suzuki, … , Mamoru Kyogashima, Takashi Takahashi
Published November 1, 2019
Citation Information: J Clin Invest. 2019;129(11):5050-5050. https://doi.org/10.1172/JCI133806.
View: Text | PDF | Amended Article

Targeting ceramide synthase 6–dependent metastasis-prone phenotype in lung cancer cells

  • Text
  • PDF
Abstract

Authors

Motoshi Suzuki, Ke Cao, Seiichi Kato, Yuji Komizu, Naoki Mizutani, Kouji Tanaka, Chinatsu Arima, Mei Chee Tai, Kiyoshi Yanagisawa, Norie Togawa, Takahiro Shiraishi, Noriyasu Usami, Tetsuo Taniguchi, Takayuki Fukui, Kohei Yokoi, Keiko Wakahara, Yoshinori Hasegawa, Yukiko Mizutani, Yasuyuki Igarashi, Jin-ichi Inokuchi, Soichiro Iwaki, Satoshi Fujii, Akira Satou, Yoko Matsumoto, Ryuichi Ueoka, Keiko Tamiya-Koizumi, Takashi Murate, Mitsuhiro Nakamura, Mamoru Kyogashima, Takashi Takahashi

×
Advertisement

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

Sign up for email alerts