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

In-Press Preview

  • 2,465 Articles
  • 0 Posts
  • ← Previous
  • 1
  • 2
  • 3
  • …
  • 246
  • 247
  • Next →
Decoding muscle-resident Schwann cell dynamics during neuromuscular junction remodeling
Steve D. Guzman, Ahmad Abu-Mahfouz, Carol S. Davis, Lloyd P. Ruiz, Peter C.D. Macpherson, Susan V. Brooks
Steve D. Guzman, Ahmad Abu-Mahfouz, Carol S. Davis, Lloyd P. Ruiz, Peter C.D. Macpherson, Susan V. Brooks
View: Text | PDF

Decoding muscle-resident Schwann cell dynamics during neuromuscular junction remodeling

  • Text
  • PDF
Abstract

This investigation leverages single-cell RNA sequencing (scRNA-Seq) to delineate the contributions of muscle-resident Schwann cells to neuromuscular junction (NMJ) remodeling by comparing a model of stable innervation with models of reinnervation following partial or complete denervation. The study discovered multiple distinct Schwann cell subtypes, including a novel terminal Schwann cell (tSC) subtype integral to the denervation-reinnervation cycle, identified by a transcriptomic signature indicative of cell migration and polarization. The data also characterizes three myelin Schwann cell subtypes, which are distinguished based on enrichment of genes associated with myelin production, mesenchymal differentiation or collagen synthesis. Importantly, SPP1 signaling emerges as a pivotal regulator of NMJ dynamics, promoting Schwann cell proliferation and muscle reinnervation across nerve injury models. These findings advance our understanding of NMJ maintenance and regeneration and underscore the therapeutic potential of targeting specific molecular pathways to treat neuromuscular and neurodegenerative disorders.

Authors

Steve D. Guzman, Ahmad Abu-Mahfouz, Carol S. Davis, Lloyd P. Ruiz, Peter C.D. Macpherson, Susan V. Brooks

×

Collagen-binding C-type natriuretic peptide enhances chondrogenesis and osteogenesis
Kenta Hirai, Kenta Sawamura, Ryusaku Esaki, Ryusuke Sawada, Yuka Okusha, Eriko Aoyama, Hiroki Saito, Kentaro Uchida, Takehiko Mima, Satoshi Kubota, Hirokazu Tsukahara, Shiro Imagama, Masaki Matsushita, Osamu Matsushita, Yasuyuki Hosono
Kenta Hirai, Kenta Sawamura, Ryusaku Esaki, Ryusuke Sawada, Yuka Okusha, Eriko Aoyama, Hiroki Saito, Kentaro Uchida, Takehiko Mima, Satoshi Kubota, Hirokazu Tsukahara, Shiro Imagama, Masaki Matsushita, Osamu Matsushita, Yasuyuki Hosono
View: Text | PDF

Collagen-binding C-type natriuretic peptide enhances chondrogenesis and osteogenesis

  • Text
  • PDF
Abstract

C-type natriuretic peptide (CNP) is known to promote chondrocyte proliferation and bone formation; however, CNP’s extremely short half-life necessitates continuous intravascular administration to achieve bone-lengthening effects. Vosoritide, a CNP analog designed for resistance to neutral endopeptidase, allows for once daily administration. Nonetheless, it distributes systemically rather than localizing to target tissues, which may result in adverse effects such as hypotension. To enhance local drug delivery and therapeutic efficacy, we developed a novel synthetic protein by fusing a collagen-binding domain (CBD) to CNP, termed CBD-CNP. This fusion protein exhibited stability under heat conditions and retained the collagen-binding ability and bioactivity as CNP. CBD-CNP localized to articular cartilage in fetal murine tibiae and promoted bone elongation. Spatial transcriptomic analysis revealed that the upregulation of chondromodulin expression may contribute to its therapeutic effects. Treatment of CBD-CNP mixed with collagen powder to a fracture site of a mouse model increased bone mineral content and bone volume rather than CNP-22. Intra-articular injection of CBD-CNP to a mouse model of knee osteoarthritis suppressed subchondral bone thickening. By addressing the limitations of CNP’s rapid degeneration, CBD-CNP leverages its collagen-binding capacity to achieve targeted, sustained delivery in collagen-rich tissues, offering a promising strategy for enhancing chondrogenesis and osteogenesis.

Authors

Kenta Hirai, Kenta Sawamura, Ryusaku Esaki, Ryusuke Sawada, Yuka Okusha, Eriko Aoyama, Hiroki Saito, Kentaro Uchida, Takehiko Mima, Satoshi Kubota, Hirokazu Tsukahara, Shiro Imagama, Masaki Matsushita, Osamu Matsushita, Yasuyuki Hosono

×

Common clonal haematopoiesis driver mutations have disparate effects on macrophage cytokines, clonal expansion and atherogenesis
Paul R. Carter, Lauren Kitt, Amanda Rodgers, Nichola Figg, Ang Zhou, Chengrui Zhu, Ziyang Wang, Peter Libby, Stephen Burgess, George S. Vassiliou, Murray CH. Clarke
Paul R. Carter, Lauren Kitt, Amanda Rodgers, Nichola Figg, Ang Zhou, Chengrui Zhu, Ziyang Wang, Peter Libby, Stephen Burgess, George S. Vassiliou, Murray CH. Clarke
View: Text | PDF

Common clonal haematopoiesis driver mutations have disparate effects on macrophage cytokines, clonal expansion and atherogenesis

  • Text
  • PDF
Abstract

Clonal haematopoiesis of indeterminate potential (CHIP) is the expansion of blood stem cells and progeny after somatic mutation. CHIP associates with increased cardiovascular disease (CVD) with inflammation from macrophages a proposed common effector. However, mouse CHIP studies are discordant for clonal expansion and inflammation. Similarly, directionality of association between CHIP and CVD remains debated. We investigated effects of three CHIP mutations on macrophage cytokines, clonal expansion and atherosclerosis in parallel. We find that Tet2 and Dnmt3a mutations increase cytokines and inflammasome activation in Tet2 but decrease in Dnmt3a. However, Jak2 mutant macrophages produced equivalent cytokine as wild-type. In mice, Tet2 mutants clonally expanded, but Dnmt3a and Jak2 mutants didn’t. Expansion was unaffected by systemic inflammation, while hyperlipidemia expanded Tet2-/- cells, but not mono-allelic mutants. Similarly, human Mendelian randomisation showed no effect of serum cytokines or CVD on CHIP risk. Experimental atherosclerosis was increased in females with Tet2 and males with Jak2, but unchanged with Dnmt3a mutations. Together, common CHIP mutations have disparate effects on macrophage cytokines and clonal expansion, and sex-dependent effects on atherogenesis, suggesting a common mechanism across CHIP is unlikely. Thus, CHIP mutations differ in pathophysiology and clinical sequalae across sexes and should be treated as different entities.

Authors

Paul R. Carter, Lauren Kitt, Amanda Rodgers, Nichola Figg, Ang Zhou, Chengrui Zhu, Ziyang Wang, Peter Libby, Stephen Burgess, George S. Vassiliou, Murray CH. Clarke

×

Health outcomes after national acute sleep deprivation events among the American public
Neil J. Kelly, Rahul Chaudhary, Wadih El Khoury, Nishita Kalepalli, Jesse Wang, Priya Patel, Irene Chan, Haris Rahman, Aisha Saiyed, Anisha N. Shah, Colleen A. McClung, Satoshi Okawa, Mehdi Nouraie, Stephen Y. Chan
Neil J. Kelly, Rahul Chaudhary, Wadih El Khoury, Nishita Kalepalli, Jesse Wang, Priya Patel, Irene Chan, Haris Rahman, Aisha Saiyed, Anisha N. Shah, Colleen A. McClung, Satoshi Okawa, Mehdi Nouraie, Stephen Y. Chan
View: Text | PDF

Health outcomes after national acute sleep deprivation events among the American public

  • Text
  • PDF
Abstract

Background: Sleep is increasingly recognized as essential to human health, yet the adverse health consequences of acute sleep deprivation are unknown. We hypothesized that acute sleep deprivation is associated with health outcomes and modulated by sleep-associated genotypes. Methods: LOESS smoothing was performed on sleep estimates from Fitbit users (N = 14,681) between June 1, 2016 and July 1, 2022. Dates when population minutes slept were less than the 90% confidence interval of the LOESS regression were named acute sleep deprivation events (ASDEs). Phenome-wide disease incidence among the AoU population (N = 287,012) in the 10 days post-ASDE was compared to a preceding reference period by McNemar test. Circadian rhythm and sleep duration-associated SNPs were screened to identify genotypes associated with shorter ASDE sleep duration. Influences of sleep and circadian genotype on post-ASDE influenza risk were modeled using binomial family generalized estimating equations. Results: We identified 32 ASDEs spanning major national events. A phenome-wide screen found increased risk of influenza (OR = 1.54 [1.40, 1.70], P-value = 1.00 x 10-18) following ASDEs. 56 SNPs were associated with decreased sleep duration on ASDEs. Higher quantiles of ASDE-related SNP genotype burden were associated with less ASDE sleep duration and a greater risk of influenza-associated healthcare visits. Conclusion: Major national events are associated with acute sleep deprivation and greater influenza risk which is amplified by sleep genotypes. These findings should inform public health vigilance surrounding major national events.

Authors

Neil J. Kelly, Rahul Chaudhary, Wadih El Khoury, Nishita Kalepalli, Jesse Wang, Priya Patel, Irene Chan, Haris Rahman, Aisha Saiyed, Anisha N. Shah, Colleen A. McClung, Satoshi Okawa, Mehdi Nouraie, Stephen Y. Chan

×

Distinct transcriptional and epigenomic programs define Hofbauer cells in term placenta
Benjámin R. Baráth, Dóra Bojcsuk, Krisztian Bene, Noemí Caballero-Sánchez, Tímea Cseh, João CR. de Freitas, Petros Tzerpos, Marta Toth, Zhonghua Tang, Seth Guller, Zoárd Tibor Krasznai, Patrícia Neuperger, Gabor J. Szebeni, Gergely Nagy, Tamás Deli, Laszlo Nagy
Benjámin R. Baráth, Dóra Bojcsuk, Krisztian Bene, Noemí Caballero-Sánchez, Tímea Cseh, João CR. de Freitas, Petros Tzerpos, Marta Toth, Zhonghua Tang, Seth Guller, Zoárd Tibor Krasznai, Patrícia Neuperger, Gabor J. Szebeni, Gergely Nagy, Tamás Deli, Laszlo Nagy
View: Text | PDF

Distinct transcriptional and epigenomic programs define Hofbauer cells in term placenta

  • Text
  • PDF
Abstract

Hofbauer cells (HBC) are fetal-derived macrophages located in the placenta that contribute to antimicrobial defense, angiogenesis, tissue remodeling, and metabolic processes within the chorionic villi. Although their roles in placental biology are increasingly recognized, the mechanisms that regulate HBC identity and function are not yet fully defined. This study aimed to define the core transcriptomic and epigenomic features of HBCs in term placentas and to examine their capacity for transcriptional responsiveness and phenotypic variation. Using chromatin accessibility profiling and bulk RNA sequencing, we found that HBCs exhibit a unique gene expression and chromatin accessibility profile compared to other fetal and adult macrophages. We identified a coordinated transcriptional network involving nuclear receptors NR4A1–3, the glucocorticoid receptor (GR), and RFX family members (RFX1, RFX2, RFX5) that appears to shape HBC identity, particularly through pathways linked to lipid metabolism and angiogenesis. Although exploratory in nature, in vitro stimulation studies showed that HBCs exhibited increased transcriptional activity in response to combined IL-4 and RSG treatment, including induction of the lipid transporter CD36. Mass cytometry analysis revealed surface markers indicative of both immature and mature macrophage states. Together, these results indicated that HBCs represent a distinct and diverse macrophage population with specialized and adaptable regulatory program in the human placenta.

Authors

Benjámin R. Baráth, Dóra Bojcsuk, Krisztian Bene, Noemí Caballero-Sánchez, Tímea Cseh, João CR. de Freitas, Petros Tzerpos, Marta Toth, Zhonghua Tang, Seth Guller, Zoárd Tibor Krasznai, Patrícia Neuperger, Gabor J. Szebeni, Gergely Nagy, Tamás Deli, Laszlo Nagy

×

Intranasal booster drives class switching and homing of memory B cells for mucosal IgA response
Si Chen, Zhengyuan Zhang, Zihan Lin, Li Yin, Lishan Ning, Wenming Liu, Qian Wang, Chenchen Yang, Bo Feng, Ying Feng, Yongping Wang, Hengchun Li, Ping He, Huan Liang, Yichu Liu, Zhixia Li, Bo Liu, Yang Li, Diana Boraschi, Linbing Qu, Xuefeng Niu, Nanshan Zhong, Pingchao Li, Ling Chen
Si Chen, Zhengyuan Zhang, Zihan Lin, Li Yin, Lishan Ning, Wenming Liu, Qian Wang, Chenchen Yang, Bo Feng, Ying Feng, Yongping Wang, Hengchun Li, Ping He, Huan Liang, Yichu Liu, Zhixia Li, Bo Liu, Yang Li, Diana Boraschi, Linbing Qu, Xuefeng Niu, Nanshan Zhong, Pingchao Li, Ling Chen
View: Text | PDF

Intranasal booster drives class switching and homing of memory B cells for mucosal IgA response

  • Text
  • PDF
Abstract

Mucosal secretory IgA (sIgA) plays a central role in protecting against the invasion of respiratory pathogen via the upper respiratory tract. To understand how intranasal booster induces mucosal sIgA response in humans, we first used liquid chromatography-tandem mass spectrometry for peptide identification of immunoglobulin (MS Ig-Seq) and single-cell B-cell receptor sequencing (scBCR-seq) to identify mucosal spike-specific sIgA monoclonal antibodies (mAbs) after intranasal booster. These mucosal sIgA mAbs exhibited enhanced neutralization up to 100-fold against SARS-CoV-2 variants compared to their monomeric IgG and IgA isotypes. Deep sequencing and longitudinal analysis of B-cell receptor repertoires revealed that nasal booster re-stimulates memory B cells primed by intramuscularly vaccination to undergo IgA class switching, somatic hypermutation, and clonal expansion. Single-cell sequencing revealed that intranasal booster upregulated the expression of mucosal homing receptors in spike-specific IgA-expressing B cells. This increase coincided with a transient increase of cytokines and chemokines that facilitate B cell recruitment in the nasal mucosa. Our findings demonstrate that intranasal booster can be an effective strategy for inducing upper respiratory mucosal sIgA and establishing mucosal immune protection.

Authors

Si Chen, Zhengyuan Zhang, Zihan Lin, Li Yin, Lishan Ning, Wenming Liu, Qian Wang, Chenchen Yang, Bo Feng, Ying Feng, Yongping Wang, Hengchun Li, Ping He, Huan Liang, Yichu Liu, Zhixia Li, Bo Liu, Yang Li, Diana Boraschi, Linbing Qu, Xuefeng Niu, Nanshan Zhong, Pingchao Li, Ling Chen

×

Insights into KIF11 pathogenesis in Microcephaly-Lymphedema-Chorioretinopathy syndrome from a lymphatic perspective
Kazim Ogmen, Sara E. Dobbins, Rose Yinghan Behncke, Ines Martinez-Corral, Ryan C.S. Brown, Michelle Meier, Sascha Ulferts, Nils Rouven Hansmeier, Ege Sackey, Ahlam Alqahtani, Christina Karapouliou, Dionysios Grigoriadis, Juan C. Del Rey Jimenez, Michael Oberlin, Denise Williams, Arzu Ekici, Kadri Karaer, Steve Jeffery, Peter Mortimer, Kristiana Gordon, Kazuhide S. Okuda, Benjamin M. Hogan, Taija Mäkinen, René Hägerling, Sahar Mansour, Silvia Martin-Almedina, Pia Ostergaard
Kazim Ogmen, Sara E. Dobbins, Rose Yinghan Behncke, Ines Martinez-Corral, Ryan C.S. Brown, Michelle Meier, Sascha Ulferts, Nils Rouven Hansmeier, Ege Sackey, Ahlam Alqahtani, Christina Karapouliou, Dionysios Grigoriadis, Juan C. Del Rey Jimenez, Michael Oberlin, Denise Williams, Arzu Ekici, Kadri Karaer, Steve Jeffery, Peter Mortimer, Kristiana Gordon, Kazuhide S. Okuda, Benjamin M. Hogan, Taija Mäkinen, René Hägerling, Sahar Mansour, Silvia Martin-Almedina, Pia Ostergaard
View: Text | PDF

Insights into KIF11 pathogenesis in Microcephaly-Lymphedema-Chorioretinopathy syndrome from a lymphatic perspective

  • Text
  • PDF
Abstract

Pathogenic variants in kinesin KIF11 underlie microcephaly-lymphedema-chorioretinopathy (MLC) syndrome. Although well known for regulating spindle dynamics ensuring successful cell division, the association of KIF11 (encoding EG5) with development of the lymphatic system, and how KIF11 pathogenic variants lead to lymphatic dysfunction and lymphedema remain unknown. Using patient-derived lymphoblastoid cells, we demonstrated that MLC patients carrying pathogenic stop-gain variants in KIF11 have reduced mRNA and protein levels. Lymphoscintigraphy showed reduced tracer absorption, and intestinal lymphangiectasia was detected in one patient, pointing to impairment of lymphatic function caused by KIF11 haploinsufficiency. We revealed that KIF11 is expressed in early human and mouse development with the lymphatic markers VEGFR3, Podoplanin and PROX1. In zebrafish, scRNA-seq identified kif11 specifically expressed in endothelial precursors. In human lymphatic endothelial cells (LECs), EG5 inhibition with Ispinesib, reduced VEGFC-driven AKT phosphorylation, migration and spheroid sprouting. KIF11 knockdown reduced PROX1 and VEGFR3 expression, providing for the first time a link between KIF11 and drivers of lymphangiogenesis and lymphatic identity.

Authors

Kazim Ogmen, Sara E. Dobbins, Rose Yinghan Behncke, Ines Martinez-Corral, Ryan C.S. Brown, Michelle Meier, Sascha Ulferts, Nils Rouven Hansmeier, Ege Sackey, Ahlam Alqahtani, Christina Karapouliou, Dionysios Grigoriadis, Juan C. Del Rey Jimenez, Michael Oberlin, Denise Williams, Arzu Ekici, Kadri Karaer, Steve Jeffery, Peter Mortimer, Kristiana Gordon, Kazuhide S. Okuda, Benjamin M. Hogan, Taija Mäkinen, René Hägerling, Sahar Mansour, Silvia Martin-Almedina, Pia Ostergaard

×

Quantitative V-gene-targeted T cell receptor sequencing as a biomarker in type 1 diabetes
Laurie G. Landry, Kristen L. Wells, Amanda M. Anderson, Kristen R. Miller, Kenneth L. Jones, Aaron W. Michels, Maki Nakayama
Laurie G. Landry, Kristen L. Wells, Amanda M. Anderson, Kristen R. Miller, Kenneth L. Jones, Aaron W. Michels, Maki Nakayama
View: Text | PDF

Quantitative V-gene-targeted T cell receptor sequencing as a biomarker in type 1 diabetes

  • Text
  • PDF
Abstract

Developing biomarkers to quantitatively monitor disease-specific T cell activity is crucial for assessing type 1 diabetes (T1D) progression and evaluating immunotherapies. This study presents an approach using V-gene targeted sequencing to quantify T cell receptor (TCR) clonotypes as biomarkers for pathogenic T cells in T1D. We identified "public" TCR clonotypes shared among multiple non-obese diabetic (NOD) mice and human organ donors, with a subset expressed exclusively by islet antigen-reactive T cells in those with T1D. Employing V-gene targeted sequencing of only TCRs containing TRAV16/16D allowed quantitative detection of the public islet antigen-reactive TCR clonotypes in peripheral blood of NOD mice. Frequencies of these public TCR clonotypes distinguished prediabetic NOD mice from those protected from diabetes. In human islets, public TCR clonotypes identical to preproinsulin-specific clones were exclusively found in T1D donors. This quantifiable TCR sequencing approach uncovered public, disease-specific clonotypes in T1D, providing biomarker candidates to monitor pathogenic T cell frequencies in blood for assessing disease activity and therapeutic response.

Authors

Laurie G. Landry, Kristen L. Wells, Amanda M. Anderson, Kristen R. Miller, Kenneth L. Jones, Aaron W. Michels, Maki Nakayama

×

Diurnal Rhythm in Chimeric Antigen Receptor T-Cell Effectiveness in an Observational Study of 715 Patients
Patrick G. Lyons, Emily Gill, Prisha Kumar, Melissa Beasley, Brenna Park-Egan, Zulfiqar A. Lokhandwala, Katie M. Lebold, Brandon Hayes-Lattin, Catherine L. Hough, Nathan Singh, Guy Hazan, Huram Mok, Janice M. Huss, Colleen A. McEvoy, Jeffrey A. Haspel
Patrick G. Lyons, Emily Gill, Prisha Kumar, Melissa Beasley, Brenna Park-Egan, Zulfiqar A. Lokhandwala, Katie M. Lebold, Brandon Hayes-Lattin, Catherine L. Hough, Nathan Singh, Guy Hazan, Huram Mok, Janice M. Huss, Colleen A. McEvoy, Jeffrey A. Haspel
View: Text | PDF

Diurnal Rhythm in Chimeric Antigen Receptor T-Cell Effectiveness in an Observational Study of 715 Patients

  • Text
  • PDF
Abstract

BACKGROUND. Chimeric antigen receptor (CAR) T-cells are a leading immunotherapy for refractory B-cell malignancies; however, their impact is limited by toxicity and incomplete efficacy. Daily (circadian) rhythms in immune function may offer a lever to boost therapeutic success; however, their clinical relevance to CAR T-cell therapy remains unknown. METHODS. We retrospectively analyzed CAR T-cell survival and complications based on infusion time at two geographically distinct hospitals in St. Louis, Missouri (n=384), and Portland, Oregon (n=331) between 1/2018 and 3/2025. The primary outcome was 90-day overall survival (OS). Secondary outcomes included event-free survival (EFS), cytokine release syndrome (CRS), immune cell-associated neurotoxicity syndrome (ICANS), ICU admission, shock, respiratory failure, and infection. We quantified the independent relationship between infusion time and outcomes using multivariable mixed-effects logistic regression and time-to-event models, adjusting for patient, oncologic, and treatment characteristics. RESULTS. The therapeutic index of CAR-T cells inversely correlated with administration time, with later infusions associated with lower effectiveness and more adverse outcomes. For each hour that CAR T-cell treatment was delayed, the adjusted odds of 90-day mortality increased by 24% (aOR 0.64-0.88, p=<0.001), severe neurotoxicity by 17% (p=0.023), and mechanical ventilation by 27% (p=0.026). These temporal patterns were most pronounced in patients receiving CD19-targeting CAR T-cell products. In contrast, we did not find an association between infusion time and severe CRS (aOR 0.99, 95% CI 0.75–1.27, p=0.92). CONCLUSION. Time of day is a potent and easily modifiable factor that could optimize CAR T-cell clinical performance. FUNDING. National Institutes of Health.

Authors

Patrick G. Lyons, Emily Gill, Prisha Kumar, Melissa Beasley, Brenna Park-Egan, Zulfiqar A. Lokhandwala, Katie M. Lebold, Brandon Hayes-Lattin, Catherine L. Hough, Nathan Singh, Guy Hazan, Huram Mok, Janice M. Huss, Colleen A. McEvoy, Jeffrey A. Haspel

×

CD8 T cells cross-restricted by HLA-B*57 and HLA-E*01 recognize HIV Gag with different functional profiles
Kevin J. Maroney, Michael A. Rose, Allisa K. Oman, Abha Chopra, Hua-Shiuan Hsieh, Zerufael Derza, Rachel Waterworth, Mark A. Brockman, Spyros A. Kalams, Anju Bansal, Paul A. Goepfert
Kevin J. Maroney, Michael A. Rose, Allisa K. Oman, Abha Chopra, Hua-Shiuan Hsieh, Zerufael Derza, Rachel Waterworth, Mark A. Brockman, Spyros A. Kalams, Anju Bansal, Paul A. Goepfert
View: Text | PDF

CD8 T cells cross-restricted by HLA-B*57 and HLA-E*01 recognize HIV Gag with different functional profiles

  • Text
  • PDF
Abstract

Few HIV-specific epitopes restricted by non-classical HLA-E have been described, and even less is known about the functional profile of responding CD8 T cells (CD8s). This study evaluates the functional characteristics of CD8s targeting the Gag epitope KF11 (KAFSPEVIPMF) restricted by either HLA-E (E-CD8s) or HLA-B57 (B57-CD8s). CD8s from eight people with HIV (PWH) were cocultured with KF11 peptide presented by cell lines expressing HLA-B*57:01, HLA-E*01:01 or E*01:03. CD8 responses were analyzed using scRNA-seq and scTCR-seq. Supernatants were also assessed for soluble protein profiling. HLA-I multimers were developed to identify CD8s restricted by HLA-B57 and/or HLA-E ex vivo. B57-CD8s secreted higher levels of cytotoxic cytokines such as IFNγ, whereas E-CD8s produced more chemotactic cytokines, including RANTES, CXCL10 (IP-10), and IL27, findings which were corroborated through scRNA sequencing. TCR clonotypes stimulated by KF11 were cross-restricted by HLA-B*57 and HLA-E*01/03 as demonstrated by in vitro T cell reporter assays and ex vivo multimer screening. Ex vivo CD8s were singly restricted by HLA-B57 and HLA-E, with dual restriction only observed in PWH with lower viral load. These findings demonstrate that certain HIV-specific CD8s in PWH exhibit dual restriction by HLA-B*57 and HLA-E*01/03, leading to functionally distinct immune responses depending upon the restricting allele(s).

Authors

Kevin J. Maroney, Michael A. Rose, Allisa K. Oman, Abha Chopra, Hua-Shiuan Hsieh, Zerufael Derza, Rachel Waterworth, Mark A. Brockman, Spyros A. Kalams, Anju Bansal, Paul A. Goepfert

×
  • ← Previous
  • 1
  • 2
  • 3
  • …
  • 246
  • 247
  • Next →

No posts were found with this tag.

Advertisement

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

Sign up for email alerts