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

Therapeutics

  • 312 Articles
  • 0 Posts
  • ← Previous
  • 1
  • 2
  • 3
  • …
  • 31
  • 32
  • Next →
Targeting intracellular LMP2 with costimulatory signal–armed antibody-like TCR T cells
Jiali Cheng, … , Taochao Tan, Liang Huang
Jiali Cheng, … , Taochao Tan, Liang Huang
Published May 22, 2025
Citation Information: JCI Insight. 2025;10(10):e178572. https://doi.org/10.1172/jci.insight.178572.
View: Text | PDF

Targeting intracellular LMP2 with costimulatory signal–armed antibody-like TCR T cells

  • Text
  • PDF
Abstract

Expanding the repertoire of CAR therapies to include intracellular antigens holds promise for treating a broad spectrum of malignancies. TCR-like T cells, capable of recognizing intracellular antigen–derived peptides in complex with HLA molecules (pHLA), represent a promising strategy in the field of engineered cellular therapy. This study introduced antibody-like TCR (abTCR) T cells that specifically targeted HLA-A*02:01–restricted LMP2426 peptides, a typical Epstein-Barr virus (EBV) latency II protein, for the treatment of EBV-associated lymphoproliferative diseases (EBV-LPDs). Compared with classic CAR T cells targeting the same epitope, abTCR T cells demonstrated superior efficiency, including increased CD107A expression, enhanced cytotoxicity, and elevated IFN-γ secretion, even when engaging with target cells that naturally present antigens. Moreover, a costimulatory signal–armed abTCR (Co-abTCR), which integrated a costimulatory structure with the abTCR, further enhanced the proliferation and in vivo tumoricidal efficacy of transfected T cells. Collectively, our study developed a potentially novel TCR-like T cell therapy that targets HLA-A*02/LMP2426 for the treatment of EBV-LPDs, providing a potential therapeutic solution for targeting of intracellular antigens in cancer immunotherapy.

Authors

Jiali Cheng, Xuelian Hu, Zhenyu Dai, Yuhao Zeng, Jin Jin, Wei Mu, Qiaoe Wei, Xiangyin Jia, Jianwei Liu, Meng Xie, Qian Luo, Guang Hu, Gaoxiang Wang, Xiaojian Zhu, Jianfeng Zhou, Min Xiao, Jue Wang, Taochao Tan, Liang Huang

×

Active synthesis of type I collagen homotrimer in Dupuytren’s fibrosis is unaffected by anti–TNF-α treatment
Kate Williamson, … , Peter D. Clegg, Elizabeth G. Canty-Laird
Kate Williamson, … , Peter D. Clegg, Elizabeth G. Canty-Laird
Published May 8, 2025
Citation Information: JCI Insight. 2025;10(9):e175188. https://doi.org/10.1172/jci.insight.175188.
View: Text | PDF

Active synthesis of type I collagen homotrimer in Dupuytren’s fibrosis is unaffected by anti–TNF-α treatment

  • Text
  • PDF
Abstract

Dupuytren’s disease is a common fibroproliferative disease of the palmar fascia of the hand, with advanced cases treated surgically. Anti-TNF injection has undergone phase 2 trials and may be effective in slowing early-stage disease progression. Here we sought to determine how new synthesis of type I collagen in Dupuytren’s differs from normal palmar fascia samples and to analyze the role of TNF in aberrant collagen synthesis. Model nonfibrotic but fibrous connective tissues were used to analyze active type I collagen protein synthesis in development, aging, and degenerative disease, where it was restricted to early development and ruptured tissue. Dupuytren’s tissue was shown to actively synthesize type I collagen, including abnormal type I collagen homotrimer. TNF-α reduced COL1A2 gene expression only in the presence of serum in 2D cell culture and had opposing effects on collagen protein production in the presence or absence of serum. TNF-α had only limited effects in 3D tendon–like constructs. Anti-TNF did not reduce type I collagen synthesis in 3D tendon–like constructs or prevent type I collagen homotrimer synthesis in Dupuytren’s tissue. Hence, modulation of the TNF-α pathway in Dupuytren’s disease is unlikely to prevent the pathological collagen accumulation that is characteristic of fibrosis.

Authors

Kate Williamson, Katie J. Lee, Emma L. Beamish, Alan Carter, Jade A. Gumbs, Gabriella Cooper, Niamh S. O’Heneghan-Yates, Lisa A. Menezes, Graham Cheung, Daniel Brown, Rob Pettitt, Brendan Geraghty, Lucy A. Bosworth, Eithne J. Comerford, Peter D. Clegg, Elizabeth G. Canty-Laird

×

The Growth Hormone-IGF-1 axis is a risk factor for Long-Term Kidney Allograft Failure
Matthew Cusick, … , Roger C. Wiggins, Abhijit S. Naik
Matthew Cusick, … , Roger C. Wiggins, Abhijit S. Naik
Published May 6, 2025
Citation Information: JCI Insight. 2025. https://doi.org/10.1172/jci.insight.188485.
View: Text | PDF

The Growth Hormone-IGF-1 axis is a risk factor for Long-Term Kidney Allograft Failure

  • Text
  • PDF
Abstract

INTRODUCTION: Maladaptive hypertrophy, podocyte stress, and depletion contribute to kidney function decline. Although IGF-1 plays a key role in early hypertrophic responses in the single kidney state, its impact on KTx outcomes remains uncertain. This report tests the hypothesis that early IGF-1 exposure reduces KTx survival. METHODS. Population datasets compared incident Death Censored Graft Failure (DCGF) rates by age at KTx (n=366,404) with IGF-1 levels by age (n=15,014). A clinical study of 216 KTx recipients evaluated the association of IGF-1 exposure with DCGF and secondary outcomes of proteinuria and Biopsy-Proven Acute Rejection. IGF-1 exposure was modeled using pre-KTx IGF-1 levels and donor kidney dose estimated from the donor:recipient body surface area ratio reflecting allograft hyperfiltration. The association of DCGF with an IGF1 SNP linked to high IGF-1 levels was assessed in 724 genotyped allograft recipients. Single-cell transcriptomic data from first-year post-KTx patients and binephric donors were compared to assess intrarenal cellular expression of IGF1, IGF1R, and GHR transcripts. RESULTS. DCGF risk by age at KTx paralleled IGF-1 levels by age. Higher IGF-1 exposure was associated with significantly increased risks of DCGF, proteinuria, and T-cell-mediated rejection. Genotypic analysis showed a 50% increase in DCGF risk per risk allele at IGF1 eQTL rs35767. First-year biopsy results revealed no increase in intrarenal IGF1 transcript, while GHR and IGF-1R transcripts were suppressed, consistent with circulating IGF-1 (vs. graft-derived IGF-1) being the primary source of IGF-1 exposure. CONCLUSION. We identify a role for the GH-IGF-1 axis in reducing KTx survival.

Authors

Matthew Cusick, Viji Nair, Damian Fermin, John Hartman, Jeffrey A. Beamish, Zeguo Sun, Zhongyang Zhang, Edgar Otto, Rajasree Menon, Sudha Nadimidla, Nicholas Demchuk, Kelly Shaffer, Peter Heeger, Weija Zhang, Madhav C. Menon, Matthias Kretzler, Roger C. Wiggins, Abhijit S. Naik

×

Biological sex and age influence GS-9620 activity ex vivo
Carissa S. Holmberg, … , Adam R. Ward, Alberto Bosque
Carissa S. Holmberg, … , Adam R. Ward, Alberto Bosque
Published May 6, 2025
Citation Information: JCI Insight. 2025. https://doi.org/10.1172/jci.insight.182242.
View: Text | PDF

Biological sex and age influence GS-9620 activity ex vivo

  • Text
  • PDF
Abstract

Toll-like receptors (TLRs) are being explored to enhance immunity in HIV cure strategies. The TLR7 agonist GS-9620 promotes immune activation, reactivates latent HIV, and delays viral rebound in some people with HIV. Previous work has shown that biological sex influences TLR7 signaling. This study examined the interplay between biological sex, age, and the sex hormones 17β-estradiol, progesterone, and testosterone on GS-9620’s ability to promote cytokine secretion and activate CD4, CD8, and NK cells ex vivo. Interestingly, sex hormones had no effect on GS-9620-mediated immune activation or cytokine induction. However, we found that GS-9620 activity was influenced by age only in female donors. Further, we found that GS-9620-mediated CD4 T cell activation was positively correlated with the induction of IFN-γ and IL-12, while CD4 T cell activation and IL-12 production were negatively correlated with age. Additionally, CD8 T cell activation was positively correlated with IFN-γ production. Mechanistically, IFN-γ was sufficient to promote higher immune activation of both CD4 and CD8 T cells in female versus male donors. In conclusion, biological sex and age, but not sex hormones, influence GS-9620-mediated immune activation. Understanding these factors will help design and evaluate future clinical trials using GS-9620 for an HIV cure.

Authors

Carissa S. Holmberg, Callie Levinger, Adam R. Ward, Alberto Bosque

×

METTL14 promotes intimal hyperplasia through m6A-mediated control of vascular smooth muscle dedifferentiation genes
Grace Chensee, … , Justin Jong Leong Wong, Renjing Liu
Grace Chensee, … , Justin Jong Leong Wong, Renjing Liu
Published April 23, 2025
Citation Information: JCI Insight. 2025. https://doi.org/10.1172/jci.insight.184444.
View: Text | PDF

METTL14 promotes intimal hyperplasia through m6A-mediated control of vascular smooth muscle dedifferentiation genes

  • Text
  • PDF
Abstract

Vascular smooth muscle cells (VSMCs) possess significant phenotypic plasticity, shifting between a contractile phenotype and a synthetic state for vascular repair/remodelling. Dysregulated VSMC transformation, marked by excessive proliferation and migration, primarily drives intimal hyperplasia. N6-methyladenosine (m6A), the most prevalent RNA modification in eukaryotes, plays a critical role in gene expression regulation; however, its impact on VSMC plasticity is not fully understood. This research investigates the alterations in m6A modification and its regulatory factors during VSMC phenotypic shifts and their influence on intimal hyperplasia. We demonstrate that METTL14, crucial for m6A deposition, significantly promotes VSMC dedifferentiation. METTL14 expression, initially negligible, is elevated in synthetic VSMC cultures, post-injury neointimal VSMCs, and human restenotic arteries. Reducing METTL14 in mouse primary VSMCs decreases pro-synthetic genes, suppressing their proliferation and migration. m6A-RIP-seq profiling shows key VSMC gene networks undergo altered m6A regulation in Mettl14-deficient cells. METTL14 enhances KLF4 and SERPINE1 expression through increased m6A deposition. Local METTL14 knockdown significantly curbs neointimal formation post-arterial injury, and reducing METTL14 in hyperplastic arteries halts further neointimal development. We found that METTL14 is a pivotal regulator of VSMC dedifferentiation, influencing KLF4- and SERPINE1-mediated phenotypic conversion. Inhibiting METTL14 is a viable strategy for preventing restenosis and halting restenotic occlusions.

Authors

Grace Chensee, Bob S.L. Lee, Immanuel D. Green, Jessica Tieng, Renhua Song, Natalia Pinello, Quintin Lee, Majid Mehravar, David A. Robinson, Mian Wang, Mary M. Kavurma, Jun Yu, Justin Jong Leong Wong, Renjing Liu

×

A small molecule PKCε inhibitor reduces hyperalgesia induced by paclitaxel or opioid withdrawal
Adriana Gregory-Flores, … , Robert O. Messing, Michela Marinelli
Adriana Gregory-Flores, … , Robert O. Messing, Michela Marinelli
Published April 22, 2025
Citation Information: JCI Insight. 2025;10(8):e186805. https://doi.org/10.1172/jci.insight.186805.
View: Text | PDF

A small molecule PKCε inhibitor reduces hyperalgesia induced by paclitaxel or opioid withdrawal

  • Text
  • PDF
Abstract

The enzyme protein kinase C ε (PKCε) plays an important role in pain signaling and represents a promising therapeutic target for the treatment of chronic pain. We designed and generated a small molecule inhibitor of PKCε, CP612, and examined its effect in a rodent model of chemotherapy-induced neuropathic pain produced by paclitaxel, which does not respond well to current therapeutics. In addition, many patients with chronic pain use opiates, which over time can become ineffective, and attempts to discontinue them can increase pain thereby promoting sustained opioid use. Therefore, we also investigated if CP612 alters pain due to opioid withdrawal. We found that CP612 attenuated hyperalgesia produced by paclitaxel, and it both prevented and reversed hyperalgesia induced by opioid withdrawal. It was not self-administered and did not affect morphine self-administration. These findings suggest that inhibition of PKCε is an effective, nonaddictive strategy to treat chemotherapy-induced neuropathic pain, with the added benefit of preventing increases in pain that occur as opioid treatment is discontinued. This latter property could benefit individuals with chronic pain who find it difficult to discontinue opioids.

Authors

Adriana Gregory-Flores, Ivan J.M. Bonet, Stève Desaivre, Jon D. Levine, Stanton F. McHardy, Harmannus C. de Kraker, Nicholas A. Clanton, Peter M. LoCoco, Nicholas M. Russell, Caleb Fleischer, Robert O. Messing, Michela Marinelli

×

CDK2 Inhibition Produces a Persistent Population of Polyploid Cancer Cells
Liliya Tyutyunyk-Massey, … , Xi Liu, Ethan Dmitrovsky
Liliya Tyutyunyk-Massey, … , Xi Liu, Ethan Dmitrovsky
Published April 15, 2025
Citation Information: JCI Insight. 2025. https://doi.org/10.1172/jci.insight.189901.
View: Text | PDF

CDK2 Inhibition Produces a Persistent Population of Polyploid Cancer Cells

  • Text
  • PDF
Abstract

Aneuploidy, a cancer hallmark, drives chromosomal instability, drug resistance, and clinically-aggressive tumors. Cyclin-dependent kinase 2 (CDK2) antagonism with independent inhibitors or CDK2 knock-down triggered anaphase catastrophe. This disrupts supernumerary centrosome clustering, causing multipolar division and apoptosis. Time-lapse fluorescent microscopy of FUCCI cell cycle probes transduced into aneuploid lung cancer cells revealed distinct fates of bipolar and polyploid cells after CDK2 inhibition. Apoptosis occurred in multipolar progeny but was repressed in persistent polyploid cancer cells. RNA-seq analyses after CDK2 inhibition of 4N versus 2N lung cancer cells were enriched for CDK1 pathway and KIF family members. The Cancer Genome Atlas (TCGA) analysis of lung cancers indicated CDK1 and KIF family member overexpression was associated with an unfavorable survival. Intravital microscopy of transplanted lung cancer cells in mice extended findings from the in vitro to in vivo settings. CDK2 inhibition of tumor-bearing mice produced polyploid cancer cells in vivo. These cancer cells were resistant to apoptosis and proliferated despite CDK2 inhibition. In contrast, polyploid populations were rarely detected in CDK2 inhibited human alveolar epithelial cells. These findings are translationally relevant. Combined targeting of CDK2 with CDK1 or kinesin family member antagonists should eliminate polyploid cancer cells, promote apoptosis and augment antineoplastic effects.

Authors

Liliya Tyutyunyk-Massey, Zibo Chen, Xiuxia Liu, Masanori Kawakami, Adam Harned, Yeap Ng, Brian Luke, Samuel C. Okpechi, Blessing Ogunlade, Yair Alfaro-Mora, Roberto Weigert, Kedar Narayan, Xi Liu, Ethan Dmitrovsky

×

CAR Treg synergy with anti-CD154 promotes infectious tolerance and dictates allogeneic heart transplant acceptance
Samarth S. Durgam, … , Megan K. Levings, Anita S. Chong
Samarth S. Durgam, … , Megan K. Levings, Anita S. Chong
Published April 8, 2025
Citation Information: JCI Insight. 2025;10(7):e188624. https://doi.org/10.1172/jci.insight.188624.
View: Text | PDF

CAR Treg synergy with anti-CD154 promotes infectious tolerance and dictates allogeneic heart transplant acceptance

  • Text
  • PDF
Abstract

Successful allograft-specific tolerance induction would eliminate the need for daily immunosuppression and improve posttransplant quality of life. Adoptive cell therapy with regulatory T cells expressing donor-specific chimeric antigen receptors (CAR Tregs) is a promising strategy but, as monotherapy, cannot prolong survival with allografts with multiple MHC mismatches. Using an HLA-A2–transgenic haplo-mismatched heart transplantation model in immunocompetent C57BL/6 recipients, we showed that HLA-A2–specific CAR (A2.CAR) Tregs were able to synergize with a low dose of anti-CD154 to enhance graft survival. Using haplo-mismatched grafts expressing the 2W-OVA transgene and tetramer-based tracking of 2W- and OVA-specific T cells, we showed that in mice with accepted grafts, A2.CAR Tregs inhibited donor-specific T cell, B cell, and antibody responses and promoted a substantial increase in endogenous FOXP3+ Tregs with indirect donor specificity. By contrast, in mice where A2.CAR Tregs failed to prolong graft survival, FOXP3– A2.CAR T cells preferentially accumulated in rejecting allografts, and endogenous donor-specific responses were not controlled. This study therefore provides evidence for synergy between A2.CAR Tregs and CD154 blockade to promote infectious tolerance in immunocompetent recipients of haplo-mismatched heart grafts and defines features of A2.CAR Tregs when they fail to reshape host immunity toward allograft tolerance.

Authors

Samarth S. Durgam, Isaac Rosado-Sánchez, Dengping Yin, Madeleine Speck, Majid Mojibian, Ismail Sayin, Grace E. Hynes, Maria-Luisa Alegre, Megan K. Levings, Anita S. Chong

×

A toxic gain of function variant in MAPK8IP3 provides novel insights into JIP3 cellular roles
Wei Zhang, … , Konstantina Skourti-Stathaki, Stanley T. Crooke
Wei Zhang, … , Konstantina Skourti-Stathaki, Stanley T. Crooke
Published March 20, 2025
Citation Information: JCI Insight. 2025. https://doi.org/10.1172/jci.insight.187199.
View: Text | PDF

A toxic gain of function variant in MAPK8IP3 provides novel insights into JIP3 cellular roles

  • Text
  • PDF
Abstract

Mitogen-activated protein kinase 8 interacting protein 3 (MAPK8IP3/JIP3) is a member of the kinesin family known to play a role in axonal transport of cargo. Mutations in the gene have been linked to severe neurodevelopmental disorders, resulting in developmental delay, intellectual disability, ataxia, tremor, autism, seizures, and visual impairment. A patient who has a missense mutation in the MAPK8IP3 gene (c. 1714 C>T, Arg578Cys) (R578C) manifests dystonia, gross motor delay and developmental delay. Here we show that the mutation is a toxic gain of function mutation which alters the interactome of JIP3, disrupts axonal transport of late endosomes, increases signaling via c-Jun N-terminal kinase (JNK), resulting in apoptosis, and disrupts the dopamine receptor 1 (D1) signaling while not affecting the dopamine receptor 2 (D2) signaling. Further, in the presence of the mutant protein, we show that 80% reduction of mutant JIP3>80% and 60% reduction of wild-type JIP3 by non-allele selective phosphorothioate (PS)-modified antisense oligonucleotides (ASOs) is well tolerated by several types of cells in vitro. Our study identifies several important new roles for JIP3 and provides important insights for therapeutic approaches, including antisense oligonucleotide reduction of JIP3.

Authors

Wei Zhang, Swapnil Mittal, Ria Thomas, Anahid Foroughishafiei, Ricardo Nunes Bastos, Wendy K. Chung, Konstantina Skourti-Stathaki, Stanley T. Crooke

×

AAV-mediated base editing restores cochlear gap junction in GJB2 dominant-negative mutation-associated syndromic hearing loss model
Takao Ukaji, … , Osamu Nureki, Kazusaku Kamiya
Takao Ukaji, … , Osamu Nureki, Kazusaku Kamiya
Published March 10, 2025
Citation Information: JCI Insight. 2025;10(5):e185193. https://doi.org/10.1172/jci.insight.185193.
View: Text | PDF

AAV-mediated base editing restores cochlear gap junction in GJB2 dominant-negative mutation-associated syndromic hearing loss model

  • Text
  • PDF
Abstract

Mutations in the gap junction β2 (GJB2) gene, which encodes connexin 26, are the leading cause of genetic deafness. These mutations are characterized by the degeneration and fragmentation of gap junctions and gap junction plaques (GJPs) composed of connexin 26. Dominant-negative mutations of GJB2, such as R75W, cause syndromic hearing loss and palmoplantar keratoderma. We previously reported that the R75W mutation, a single-base substitution where C is replaced by T, causes fragmentation of GJPs. Therefore, an adenine base editor (ABE), which enables A-to-G base conversions, can potentially be useful for the treatment of this genetic disease. Here, we report that an all-in-one adeno-associated virus (AAV) vector, which includes a compact ABE (SaCas9-NNG-ABE8e) with broad targeting range, and a sgRNA targeting the R75W mutation in GJB2 corrected this pathogenic mutation and facilitated the recovery of the gap junction intercellular communication network of GJPs. In a transgenic mouse model with the GJB2 R75W mutation, AAV-mediated base editing also restored the fragmented GJPs to orderly outlines in cochlear supporting cells. Our findings suggest that an ABE-based base-editing strategy could be an optimal treatment for the dominant form of GJB2-related hearing loss, GJB2-related skin diseases, and other deafness-related mutations, especially single-base substitutions.

Authors

Takao Ukaji, Daisuke Arai, Harumi Tsutsumi, Ryoya Nakagawa, Fumihiko Matsumoto, Katsuhisa Ikeda, Osamu Nureki, Kazusaku Kamiya

×
  • ← Previous
  • 1
  • 2
  • 3
  • …
  • 31
  • 32
  • Next →

No posts were found with this tag.

Advertisement

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

Sign up for email alerts