Go to The Journal of Clinical Investigation
  • About
  • Editors
  • Consulting Editors
  • For authors
  • Publication ethics
  • Publication alerts by email
  • Transfers
  • Advertising
  • Job board
  • Contact
  • Physician-Scientist Development
  • Current issue
  • Past issues
  • By specialty
    • COVID-19
    • Cardiology
    • Immunology
    • Metabolism
    • Nephrology
    • Oncology
    • Pulmonology
    • All ...
  • Videos
  • Collections
    • In-Press Preview
    • Resource and Technical Advances
    • Clinical Research and Public Health
    • Research Letters
    • Editorials
    • Perspectives
    • Physician-Scientist Development
    • Reviews
    • Top read articles

  • Current issue
  • Past issues
  • Specialties
  • In-Press Preview
  • Resource and Technical Advances
  • Clinical Research and Public Health
  • Research Letters
  • Editorials
  • Perspectives
  • Physician-Scientist Development
  • Reviews
  • Top read articles
  • About
  • Editors
  • Consulting Editors
  • For authors
  • Publication ethics
  • Publication alerts by email
  • Transfers
  • Advertising
  • Job board
  • Contact

Immunology

  • 1,084 Articles
  • 0 Posts
  • ← Previous
  • 1
  • 2
  • …
  • 89
  • 90
  • 91
  • …
  • 108
  • 109
  • Next →
Unexpected kidney-restricted role for IL-17 receptor signaling in defense against systemic Candida albicans infection
Kritika Ramani, … , Jay K. Kolls, Partha S. Biswas
Kritika Ramani, … , Jay K. Kolls, Partha S. Biswas
Published May 3, 2018
Citation Information: JCI Insight. 2018;3(9):e98241. https://doi.org/10.1172/jci.insight.98241.
View: Text | PDF

Unexpected kidney-restricted role for IL-17 receptor signaling in defense against systemic Candida albicans infection

  • Text
  • PDF
Abstract

Kidney injury is a frequent outcome in patients with disseminated Candida albicans fungal infections. IL-17 receptor (IL-17R) signaling is critical for renal protection against disseminated candidiasis, but the identity and function of IL-17–responsive cells in mediating renal defense remains an active area of debate. Using BM chimeras, we found that IL-17R signaling is required only in nonhematopoietic cells for immunity to systemic C. albicans infection. Since renal tubular epithelial cells (RTEC) are highly responsive to IL-17 in vitro, we hypothesized that RTEC might be the dominant target of IL-17 activity in the infected kidney. We generated mice with a conditional deletion of IL-17 receptor A (Il17ra) in RTEC (Il17raΔRTEC). Strikingly, Il17raΔRTEC mice showed enhanced kidney damage and early mortality following systemic infection, very similar to Il17ra–/– animals. Increased susceptibility to candidiasis in Il17raΔRTEC mice was associated with diminished activation of the renal protective Kallikrein-kinin system (KKS), resulting in reduced apoptosis of kidney-resident cells during hyphal invasion. Moreover, protection was restored by treatment with bradykinin, the major end-product of KKS activation, which was mediated dominantly via bradykinin receptor b1. These data show that IL-17R signaling in RTEC is necessary and likely sufficient for IL-17–mediated renal defense against fatal systemic C. albicans infection.

Authors

Kritika Ramani, Chetan V. Jawale, Akash H. Verma, Bianca M. Coleman, Jay K. Kolls, Partha S. Biswas

×

First-in-human topical microbiome transplantation with Roseomonas mucosa for atopic dermatitis
Ian A. Myles, … , Gulbu Uzel, Sandip K. Datta
Ian A. Myles, … , Gulbu Uzel, Sandip K. Datta
Published May 3, 2018
Citation Information: JCI Insight. 2018;3(9):e120608. https://doi.org/10.1172/jci.insight.120608.
View: Text | PDF

First-in-human topical microbiome transplantation with Roseomonas mucosa for atopic dermatitis

  • Text
  • PDF
Abstract

The underlying pathology of atopic dermatitis (AD) includes impaired skin barrier function, susceptibility to Staphylococcus aureus skin infection, immune dysregulation, and cutaneous dysbiosis. Our recent investigation into the potential role of Gram-negative skin bacteria in AD revealed that isolates of one particular commensal, Roseomonas mucosa, collected from healthy volunteers (HVs) improved outcomes in mouse and cell culture models of AD. In contrast, isolates of R. mucosa from patients with AD worsened outcomes in these models. These preclinical results suggested that interventions targeting the microbiome could provide therapeutic benefit for patients with AD. As a first test of this hypothesis in humans, 10 adult and 5 pediatric patients were enrolled in an open-label phase I/II safety and activity trial (the Beginning Assessment of Cutaneous Treatment Efficacy for Roseomonas in Atopic Dermatitis trial; BACTERiAD I/II). Treatment with R. mucosa was associated with significant decreases in measures of disease severity, topical steroid requirement, and S. aureus burden. There were no adverse events or treatment complications. We additionally evaluated differentiating bacterial metabolites and topical exposures that may contribute to the skin dysbiosis associated with AD and/or influence future microbiome-based treatments. These early results support continued evaluation of R. mucosa therapy with a placebo-controlled trial.

Authors

Ian A. Myles, Noah J. Earland, Erik D. Anderson, Ian N. Moore, Mark D. Kieh, Kelli W. Williams, Arhum Saleem, Natalia M. Fontecilla, Pamela A. Welch, Dirk A. Darnell, Lisa A. Barnhart, Ashleigh A. Sun, Gulbu Uzel, Sandip K. Datta

×

CX3CR1 identifies PD-1 therapy–responsive CD8+ T cells that withstand chemotherapy during cancer chemoimmunotherapy
Yiyi Yan, … , Roxana S. Dronca, Haidong Dong
Yiyi Yan, … , Roxana S. Dronca, Haidong Dong
Published April 19, 2018
Citation Information: JCI Insight. 2018;3(8):e97828. https://doi.org/10.1172/jci.insight.97828.
View: Text | PDF

CX3CR1 identifies PD-1 therapy–responsive CD8+ T cells that withstand chemotherapy during cancer chemoimmunotherapy

  • Text
  • PDF
Abstract

Although immune checkpoint inhibitors have resulted in durable clinical benefits in a subset of patients with advanced cancer, some patients who did not respond to initial anti–PD-1 therapy have been found to benefit from the addition of salvage chemotherapy. However, the mechanism responsible for the successful chemoimmunotherapy is not completely understood. Here we show that a subset of circulating CD8+ T cells expressing the chemokine receptor CX3CR1 are able to withstand the toxicity of chemotherapy and are increased in patients with metastatic melanoma who responded to chemoimmunotherapy (paclitaxel and carboplatin plus PD-1 blockade). These CX3CR1+CD8+ T cells have effector memory phenotypes and the ability to efflux chemotherapy drugs via the ABCB1 transporter. In line with clinical observation, our preclinical models identified an optimal sequencing of chemoimmunotherapy that resulted in an increase of CX3CR1+CD8+ T cells. Taken together, we found a subset of PD-1 therapy–responsive CD8+ T cells that were capable of withstanding chemotherapy and executing tumor rejection with their unique abilities of drug efflux (ABCB1), cytolytic activity (granzyme B and perforin), and migration to and retention (CX3CR1 and CD11a) at tumor sites. Future strategies to monitor and increase the frequency of CX3CR1+CD8+ T cells may help to design effective chemoimmunotherapy to overcome cancer resistance to immune checkpoint blockade therapy.

Authors

Yiyi Yan, Siyu Cao, Xin Liu, Susan M. Harrington, Wendy E. Bindeman, Alex A. Adjei, Jin Sung Jang, Jin Jen, Ying Li, Pritha Chanana, Aaron S. Mansfield, Sean S. Park, Svetomir N. Markovic, Roxana S. Dronca, Haidong Dong

×

Engineered T cells targeting E7 mediate regression of human papillomavirus cancers in a murine model
Benjamin Y. Jin, … , Cornelia L. Trimble, Christian S. Hinrichs
Benjamin Y. Jin, … , Cornelia L. Trimble, Christian S. Hinrichs
Published April 19, 2018
Citation Information: JCI Insight. 2018;3(8):e99488. https://doi.org/10.1172/jci.insight.99488.
View: Text | PDF

Engineered T cells targeting E7 mediate regression of human papillomavirus cancers in a murine model

  • Text
  • PDF
Abstract

T cell receptor (TCR) T cell therapy is a promising cancer treatment modality. However, its successful development for epithelial cancers may depend on the identification of high-avidity TCRs directed against tumor-restricted target antigens. The human papillomavirus (HPV) E7 antigen is an attractive therapeutic target that is constitutively expressed by HPV+ cancers but not by healthy tissues. It is unknown if genetically engineered TCR T cells that target E7 can mediate regression of HPV+ cancers. We identified an HPV-16 E7-specific, HLA-A*02:01-restricted TCR from a uterine cervix biopsy from a woman with cervical intraepithelial neoplasia. This TCR demonstrated high functional avidity, with CD8 coreceptor–independent tumor targeting. Human T cells transduced to express the TCR specifically recognized and killed HPV-16+ cervical and oropharyngeal cancer cell lines and mediated regression of established HPV-16+ human cervical cancer tumors in a mouse model. These findings support the therapeutic potential of this approach and established the basis for an E7 TCR gene therapy clinical trial in patients with metastatic HPV+ cancers (NCT02858310).

Authors

Benjamin Y. Jin, Tracy E. Campbell, Lindsey M. Draper, Sanja Stevanović, Bianca Weissbrich, Zhiya Yu, Nicholas P. Restifo, Steven A. Rosenberg, Cornelia L. Trimble, Christian S. Hinrichs

×

CXCL10 stabilizes T cell–brain endothelial cell adhesion leading to the induction of cerebral malaria
Elizabeth W. Sorensen, … , Thorsten R. Mempel, Andrew D. Luster
Elizabeth W. Sorensen, … , Thorsten R. Mempel, Andrew D. Luster
Published April 19, 2018
Citation Information: JCI Insight. 2018;3(8):e98911. https://doi.org/10.1172/jci.insight.98911.
View: Text | PDF

CXCL10 stabilizes T cell–brain endothelial cell adhesion leading to the induction of cerebral malaria

  • Text
  • PDF
Abstract

Malaria remains one of the world’s most significant human infectious diseases and cerebral malaria (CM) is its most deadly complication. CM pathogenesis remains incompletely understood, hindering the development of therapeutics to prevent this lethal complication. Elevated levels of the chemokine CXCL10 are a biomarker for CM, and CXCL10 and its receptor CXCR3 are required for experimental CM (ECM) in mice, but their role has remained unclear. Using multiphoton intravital microscopy, CXCR3 receptor– and ligand–deficient mice and bone marrow chimeric mice, we demonstrate a key role for endothelial cell–produced CXCL10 in inducing the firm adhesion of T cells and preventing their cell detachment from the brain vasculature. Using a CXCL9 and CXCL10 dual-CXCR3-ligand reporter mouse, we found that CXCL10 was strongly induced in the brain endothelium as early as 4 days after infection, while CXCL9 and CXCL10 expression was found in inflammatory monocytes and monocyte-derived DCs within the blood vasculature on day 8. The induction of both CXCL9 and CXCL10 was completely dependent on IFN-γ receptor signaling. These data demonstrate that IFN-γ–induced, endothelium-derived CXCL10 plays a critical role in mediating the T cell–endothelial cell adhesive events that initiate the inflammatory cascade that injures the endothelium and induces the development of ECM.

Authors

Elizabeth W. Sorensen, Jeffrey Lian, Aleksandra J. Ozga, Yoshishige Miyabe, Sophina W. Ji, Shannon K. Bromley, Thorsten R. Mempel, Andrew D. Luster

×

Whole-exome sequencing uncovers oxidoreductases DHTKD1 and OGDHL as linkers between mitochondrial dysfunction and eosinophilic esophagitis
Joseph D. Sherrill, … , Taosheng Huang, Marc E. Rothenberg
Joseph D. Sherrill, … , Taosheng Huang, Marc E. Rothenberg
Published April 19, 2018
Citation Information: JCI Insight. 2018;3(8):e99922. https://doi.org/10.1172/jci.insight.99922.
View: Text | PDF

Whole-exome sequencing uncovers oxidoreductases DHTKD1 and OGDHL as linkers between mitochondrial dysfunction and eosinophilic esophagitis

  • Text
  • PDF
Abstract

Eosinophilic esophagitis (EoE) is an allergic inflammatory esophageal disorder with a complex underlying genetic etiology often associated with other comorbidities. Using whole-exome sequencing (WES) of 63 patients with EoE and 60 unaffected family members and family-based trio analysis, we sought to uncover rare coding variants. WES analysis identified 5 rare, damaging variants in dehydrogenase E1 and transketolase domain–containing 1 (DHTKD1). Rare variant burden analysis revealed an overabundance of putative, potentially damaging DHTKD1 mutations in EoE (P = 0.01). Interestingly, we also identified 7 variants in the DHTKD1 homolog oxoglutarate dehydrogenase-like (OGDHL). Using shRNA-transduced esophageal epithelial cells and/or patient fibroblasts, we further showed that disruption of normal DHTKD1 or OGDHL expression blunts mitochondrial function. Finally, we demonstrated that the loss of DHTKD1 expression increased ROS production and induced the expression of viperin, a gene previously shown to be involved in production of Th2 cytokines in T cells. Viperin had increased expression in esophageal biopsies of EoE patients compared with control individuals and was upregulated by IL-13 in esophageal epithelial cells. These data identify a series of rare genetic variants implicating DHTKD1 and OGDHL in the genetic etiology of EoE and underscore a potential pathogenic role for mitochondrial dysfunction in EoE.

Authors

Joseph D. Sherrill, Kiran KC, Xinjian Wang, Ting Wen, Adam Chamberlin, Emily M. Stucke, Margaret H. Collins, J. Pablo Abonia, Yanyan Peng, Qiang Wu, Philip E. Putnam, Phillip J. Dexheimer, Bruce J. Aronow, Leah C. Kottyan, Kenneth M. Kaufman, John B. Harley, Taosheng Huang, Marc E. Rothenberg

×

The inflammasome potentiates influenza/Staphylococcus aureus superinfection in mice
Keven M. Robinson, … , Jieru Wang, John F. Alcorn
Keven M. Robinson, … , Jieru Wang, John F. Alcorn
Published April 5, 2018
Citation Information: JCI Insight. 2018;3(7):e97470. https://doi.org/10.1172/jci.insight.97470.
View: Text | PDF

The inflammasome potentiates influenza/Staphylococcus aureus superinfection in mice

  • Text
  • PDF
Abstract

Secondary bacterial respiratory infections are commonly associated with both acute and chronic lung injury. Influenza complicated by bacterial pneumonia is an effective model to study host defense during pulmonary superinfection due to its clinical relevance. Multiprotein inflammasomes are responsible for IL-1β production in response to infection and drive tissue inflammation. In this study, we examined the role of the inflammasome during viral/bacterial superinfection. We demonstrate that ASC–/– mice are protected from bacterial superinfection and produce sufficient quantities of IL-1β through an apoptosis-associated speck-like protein containing CARD (ASC) inflammasome–independent mechanism. Despite the production of IL-1β by ASC–/– mice in response to bacterial superinfection, these mice display decreased lung inflammation. A neutrophil elastase inhibitor blocked ASC inflammasome–independent production of IL-1β and the IL-1 receptor antagonist, anakinra, confirmed that IL-1 remains crucial to the clearance of bacteria during superinfection. Delayed inhibition of NLRP3 during influenza infection by MCC950 decreases bacterial burden during superinfection and leads to decreased inflammatory cytokine production. Collectively, our results demonstrate that ASC augments the clearance of bacteria, but can also contribute to inflammation and mortality. ASC should be considered as a therapeutic target to decrease morbidity and mortality during bacterial superinfection.

Authors

Keven M. Robinson, Krishnaveni Ramanan, Michelle E. Clay, Kevin J. McHugh, Matthew J. Pilewski, Kara L. Nickolich, Catherine Corey, Sruti Shiva, Jieru Wang, John F. Alcorn

×

IL-27 gene therapy induces depletion of Tregs and enhances the efficacy of cancer immunotherapy
Jianmin Zhu, … , Yang Liu, Xue-Feng Bai
Jianmin Zhu, … , Yang Liu, Xue-Feng Bai
Published April 5, 2018
Citation Information: JCI Insight. 2018;3(7):e98745. https://doi.org/10.1172/jci.insight.98745.
View: Text | PDF

IL-27 gene therapy induces depletion of Tregs and enhances the efficacy of cancer immunotherapy

  • Text
  • PDF
Abstract

Tumor-induced expansion of Tregs is a significant obstacle to cancer immunotherapy. However, traditional approaches to deplete Tregs are often inefficient, provoking autoimmunity. We show here that administration of IL-27–expressing recombinant adeno-associated virus (AAV–IL-27) significantly inhibits tumor growth and enhances T cell responses in tumors. Strikingly, we found that AAV–IL-27 treatment causes rapid depletion of Tregs in peripheral blood, lymphoid organs, and — most pronouncedly — tumor microenvironment. AAV–IL-27–mediated Treg depletion is dependent on IL-27 receptor and Stat1 in Tregs and is a combined result of CD25 downregulation in Tregs and inhibition of IL-2 production by T cells. In combination with a GM-CSF vaccine, AAV–IL-27 treatment not only induced nearly complete tumor rejection, but also resulted in amplified neoantigen-specific T cell responses. AAV–IL-27 also dramatically increased the efficacy of anti–PD-1 therapy, presumably due to induction of PD-L1 in T cells and depletion of Tregs. Importantly, AAV–IL-27 therapy did not induce significant adverse events, partially due to its induction of IL-10. In a plasmacytoma mouse model, we found that IL-10 was required for AAV–IL-27–mediated tumor rejection. Thus, our study demonstrates the potential of AAV–IL-27 as an independent cancer therapeutic and as an efficient adjuvant for cancer immunotherapy.

Authors

Jianmin Zhu, Jin-Qing Liu, Min Shi, Xinhua Cheng, Miao Ding, Jianchao C. Zhang, Jonathan P. Davis, Sanjay Varikuti, Abhay R. Satoskar, Lanchun Lu, Xueliang Pan, Pan Zheng, Yang Liu, Xue-Feng Bai

×

Comprehensive immunoproteogenomic analyses of malignant pleural mesothelioma
Hyun-Sung Lee, … , David J. Sugarbaker, Bryan M. Burt
Hyun-Sung Lee, … , David J. Sugarbaker, Bryan M. Burt
Published April 5, 2018
Citation Information: JCI Insight. 2018;3(7):e98575. https://doi.org/10.1172/jci.insight.98575.
View: Text | PDF

Comprehensive immunoproteogenomic analyses of malignant pleural mesothelioma

  • Text
  • PDF
Abstract

We generated a comprehensive atlas of the immunologic cellular networks within human malignant pleural mesothelioma (MPM) using mass cytometry. Data-driven analyses of these high-resolution single-cell data identified 2 distinct immunologic subtypes of MPM with vastly different cellular composition, activation states, and immunologic function; mass spectrometry demonstrated differential abundance of MHC-I and -II neopeptides directly identified between these subtypes. The clinical relevance of this immunologic subtyping was investigated with a discriminatory molecular signature derived through comparison of the proteomes and transcriptomes of these 2 immunologic MPM subtypes. This molecular signature, representative of a favorable intratumoral cell network, was independently associated with improved survival in MPM and predicted response to immune checkpoint inhibitors in patients with MPM and melanoma. These data additionally suggest a potentially novel mechanism of response to checkpoint blockade: requirement for high measured abundance of neopeptides in the presence of high expression of MHC proteins specific for these neopeptides.

Authors

Hyun-Sung Lee, Hee-Jin Jang, Jong Min Choi, Jun Zhang, Veronica Lenge de Rosen, Thomas M. Wheeler, Ju-Seog Lee, Thuydung Tu, Peter T. Jindra, Ronald H. Kerman, Sung Yun Jung, Farrah Kheradmand, David J. Sugarbaker, Bryan M. Burt

×

Human MAIT cells exit peripheral tissues and recirculate via lymph in steady state conditions
Valentin Voillet, … , Michael R. Betts, Martin Prlic
Valentin Voillet, … , Michael R. Betts, Martin Prlic
Published April 5, 2018
Citation Information: JCI Insight. 2018;3(7):e98487. https://doi.org/10.1172/jci.insight.98487.
View: Text | PDF

Human MAIT cells exit peripheral tissues and recirculate via lymph in steady state conditions

  • Text
  • PDF
Abstract

Mucosal-associated invariant T cells (MAIT cells) recognize bacterial metabolites as antigen and are found in blood and tissues, where they are poised to contribute to barrier immunity. Recent data demonstrate that MAIT cells located in mucosal barrier tissues are functionally distinct from their blood counterparts, but the relationship and circulation of MAIT cells between blood and different tissue compartments remains poorly understood. Previous studies raised the possibility that MAIT cells do not leave tissue and may either be retained or undergo apoptosis. To directly address if human MAIT cells exit tissues, we collected human donor–matched thoracic duct lymph and blood and analyzed MAIT cell phenotype, transcriptome, and T cell receptor (TCR) diversity by flow cytometry and RNA sequencing. We found that MAIT cells were present in the lymph, despite being largely CCR7– in the blood, thus indicating that MAIT cells in the lymph migrated from tissues and were capable of exiting tissues to recirculate. Importantly, MAIT cells in the lymph and blood had highly overlapping clonotype usage but distinct transcriptome signatures, indicative of differential activation states.

Authors

Valentin Voillet, Marcus Buggert, Chloe K. Slichter, Julia D. Berkson, Florian Mair, Mary M. Addison, Yoav Dori, Gregory Nadolski, Maxim G. Itkin, Raphael Gottardo, Michael R. Betts, Martin Prlic

×
  • ← Previous
  • 1
  • 2
  • …
  • 89
  • 90
  • 91
  • …
  • 108
  • 109
  • Next →

No posts were found with this tag.

Advertisement

Copyright © 2025 American Society for Clinical Investigation
ISSN 2379-3708

Sign up for email alerts