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

Nephrology

  • 268 Articles
  • 0 Posts
  • ← Previous
  • 1
  • 2
  • 3
  • …
  • 26
  • 27
  • Next →
NaCl and urea modulate CD8+ T cell survival, renal accumulation and response to BK virus
Peyman Falahat, … , Marieta Toma, Sibylle von Vietinghoff
Peyman Falahat, … , Marieta Toma, Sibylle von Vietinghoff
Published August 26, 2025
Citation Information: JCI Insight. 2025. https://doi.org/10.1172/jci.insight.194570.
View: Text | PDF

NaCl and urea modulate CD8+ T cell survival, renal accumulation and response to BK virus

  • Text
  • PDF
Abstract

BK virus nephropathy is a severe, graft-threatening complication of kidney transplantation that requires an effective T cell response. It typically emerges in the kidney medulla. Elevated osmolyte concentrations that dynamically respond to loop diuretic therapy characterize this environment. BK-viremia development in kidney graft recipients negatively correlated with loop diuretic therapy. The association remained significant in multivariable and propensity score matched analyses. Kidney function was better preserved and CD8+ T cell abundance higher in loop diuretic-exposed allografts. CD8+ T cell densities in healthy human and murine kidney medulla were lower than in cortex and increased upon loop diuretic therapy in mice. As a potential underlying mechanism, kidney medullary NaCl and urea concentrations decreased primary human CD8+ T cell numbers in vitro by induction of cell death and limitation of proliferation, respectively. Both osmolytes downregulated interferon-related gene expression. NaCl induced p53-dependent apoptosis and upregulated Na+-transporter SLC38A2, which promoted caspase 3 activation. Both decreased T cell response and cytokine secretion in response to viral peptide and allogenic tubular epithelial cell killing, components of anti-BKV response in the kidney allograft. Our results propose osmolyte-mediated mitigation of CD8+ T cell function as a what we believe to be novel mechanism that impairs immune response to BK virus, therapeutic potential of which is testable.

Authors

Peyman Falahat, Adrian Goldspink, Lucia Oehler, Jessica Schmitz, Julia Miranda, Islem Gammoudi, Jan Hinrich Bräsen, Niklas Klümper, Olena Babyak, Christian Kurts, Herrmann Haller, Marieta Toma, Sibylle von Vietinghoff

×

The spleen tyrosine kinase inhibitor entospletinib resolves inflammation to promote repair following acute kidney injury
Esteban E. Elias, … , Justin Chun, Daniel A. Muruve
Esteban E. Elias, … , Justin Chun, Daniel A. Muruve
Published August 22, 2025
Citation Information: JCI Insight. 2025;10(16):e189601. https://doi.org/10.1172/jci.insight.189601.
View: Text | PDF
Article has an altmetric score of 3

The spleen tyrosine kinase inhibitor entospletinib resolves inflammation to promote repair following acute kidney injury

  • Text
  • PDF
Abstract

Nonresolving inflammation and maladaptive renal repair contribute to the pathogenesis of acute kidney injury (AKI) transition to chronic kidney disease (CKD). Few therapies have been identified that can modulate these injurious pathways following AKI. Spleen tyrosine kinase (SYK) is an immune regulator expressed in the kidney and a potential therapeutic target for AKI. The effect of the selective SYK inhibitor entospletinib was studied in AKI-to-CKD transition. Entospletinib was administered to mice undergoing unilateral renal ischemia-reperfusion injury (IRI), with kidneys analyzed over 14 days. Single-cell RNA sequencing, digital spatial profiling, intravital microscopy, and flow cytometry were employed to study renal phenotypes. Entospletinib administered before and after IRI protected ischemic kidneys and significantly attenuated the transition to CKD. Entospletinib targeted leukocyte-expressed SYK and prevented neutrophil/monocyte recruitment to the kidney. Entospletinib reduced nonresolving tubulointerstitial inflammation after AKI by blocking activation of mannose receptor-1– and C-type lectin domain family 7 member A–expressing proinflammatory macrophages. The resolution of renal inflammation mediated by entospletinib was associated with a reciprocal increase in resident macrophages, reparative gene expression, preserved tubular integrity, and reduced renal fibrosis. The SYK inhibitor entospletinib resolves renal inflammation and promotes repair following AKI.

Authors

Esteban E. Elias, Arthur Lau, Sisay Getie Belay, Afshin Derakhshani, Graciela Andonegui, Craig N. Jenne, Antoine Dufour, Nathan A. Bracey, Justin Chun, Daniel A. Muruve

×

Urine Proteomic Signatures of Kidney Function Decline after Hospitalization
Yumeng Wen, … , Jonathan Himmelfarb, Chirag R. Parikh
Yumeng Wen, … , Jonathan Himmelfarb, Chirag R. Parikh
Published August 12, 2025
Citation Information: JCI Insight. 2025. https://doi.org/10.1172/jci.insight.195577.
View: Text | PDF
Article has an altmetric score of 1

Urine Proteomic Signatures of Kidney Function Decline after Hospitalization

  • Text
  • PDF
Abstract

BACKGROUND. Urine proteomics may provide mechanistic insights on why patients experience a higher risk of kidney function decline after hospitalization. METHDOS. In 174 patients with and without acute kidney injury (AKI) from the Assessment, Serial Evaluation, and Subsequent Sequelae in AKI (ASSESS-AKI) cohort, we used Olink to profile 2783 urine proteins collected at 3 months post-hospitalization and determined their association with estimated glomerular filtration rate (eGFR) decline during median [IQR] of 5.1[4.0-6.0] years follow-up. In four independent cohorts including the Kidney Precision Medicine Project (KPMP), we determined if proteins were differentially expressed with AKI. We used weighted correlation network analysis to determine proteins’ cellular enrichment in the kidney transcriptome (single-cell and spatial transcriptomics) in patients with AKI receiving research kidney biopsy. RESULTS. We identified 387 and 10 proteins associated with faster and slower eGFR decline, respectively, most of which were differentially expressed in patients at the time of AKI. Among these proteins, 283 (71%) were expressed by kidney cells in participants with AKI from KPMP. The expression formed 3 clusters enriched in the proximal tubule, degenerative tubule and myeloid cells, and stromal cells, and correlated with histopathological features of AKI, such as tubular injury, interstitial inflammation, and fibrosis, respectively. CONCLUSION. Urinary proteins reflecting degenerative tubular injury, inflammation, and fibrosis are associated with eGFR decline in recently hospitalized patients. FUNDING. The Kidney Precision Medicine Project (KPMP) is supported by the National Institute of Diabetes and Digestive Kidney Diseases (NIDDK) through the following grantsU01DK133081, U01DK133091, U01DK133092, U01DK133093, U01DK133095, U01DK133097, U01DK114866, U01DK114908, U01DK133090, U01DK133113, U01DK133766, U01DK133768, U01DK114907, U01DK114920, U01DK114923, U01DK114933, U24DK114886, UH3DK114926, UH3DK114861, UH3DK114915, and UH3DK114937 We gratefully acknowledge the essential contributions of our patient participants and support of the American public though their tax dollars. SM is supported by NIDDK Grant K23DK128358.

Authors

Yumeng Wen, Steven Menez, Heather Thiessen Philbrook, Dennis Moledina, Steven G. Coca, Jiashu Xue, James Kaufman, Vernon Chinchillil, Paul L. Kimmel, T. Alp Ikizler, Chi-yuan Hsu, Tanika Kelly, Ana Ricardo, Jonathan Himmelfarb, Chirag R. Parikh

×

Transgenic augmentation of erythroferrone in mice ameliorates anemia in adenine-induced chronic kidney disease
Brian Czaya, … , Elizabeta Nemeth, Tomas Ganz
Brian Czaya, … , Elizabeta Nemeth, Tomas Ganz
Published August 7, 2025
Citation Information: JCI Insight. 2025. https://doi.org/10.1172/jci.insight.190018.
View: Text | PDF

Transgenic augmentation of erythroferrone in mice ameliorates anemia in adenine-induced chronic kidney disease

  • Text
  • PDF
Abstract

Anemia is a common and disabling complication of chronic kidney disease (CKD). Current therapies can be burdensome, and full correction of anemia is limited by cardiovascular side effects. New approaches that may offer additional therapeutic options are needed. We explored the anti-anemic effects of erythroferrone, an erythroid hormone that induces iron mobilization by suppressing the master iron-regulatory hormone hepcidin. In a preclinical murine model of adenine-induced CKD, transgenic augmentation of erythroferrone mobilized iron, increased hemoglobin concentrations by approximately 2 g/dl, and modestly improved renal function without affecting systemic or renal inflammation, fibrosis, or markers of mineral metabolism. This study supports the concept that therapeutic augmentation of erythroferrone is a promising approach for alleviating CKD-associated anemia.

Authors

Brian Czaya, Joseph D. Olivera, Moya Zhang, Amber Lundin, Christian D. Castro Andrade, Grace Jung, Mark R. Hanudel, Elizabeta Nemeth, Tomas Ganz

×

Hyperosmotic stimuli activate polycystin proteins to aid in urine concentration
Karla M. Márquez-Nogueras, … , Darren P. Wallace, Ivana Y. Kuo
Karla M. Márquez-Nogueras, … , Darren P. Wallace, Ivana Y. Kuo
Published August 5, 2025
Citation Information: JCI Insight. 2025. https://doi.org/10.1172/jci.insight.186290.
View: Text | PDF
Article has an altmetric score of 3

Hyperosmotic stimuli activate polycystin proteins to aid in urine concentration

  • Text
  • PDF
Abstract

Autosomal dominant polycystic kidney disease (ADPKD) is caused by mutations in PKD1 or PKD2, which encode polycystin-1 (PC1) and polycystin-2 (PC2) respectively. These proteins are thought to form a signaling complex that can flux cations including calcium. One of the earliest symptoms in ADPKD is a decline in the concentrating ability of the kidneys, occurring prior to cyst formation. We reasoned that hyperosmolality stimulates the polycystin complex, and that the loss of this function impairs water reabsorption. We found that hyperosmolality resulted in the phosphorylation of a microtubule associated protein 4 (MAP4) in a PC1-dependent manner which then elicited ER-localized PC2 calcium signals. ER-localized PC2 hyperosmotic calcium signals were required for trafficking of the water channel aquaporin (AQP2). Pre-cystic PC1-KO and PC2-KO murine kidneys had cytosolic localized AQP2, and diluted urine compared to their respective controls. Kidney tissue sections from ADPKD patients showed decreased AQP2 apical membrane localization in cystic and non-cystic tubules. Our study demonstrates that osmolality is a physiological stimulus of the polycystin complex, and loss of polycystin osmosensing results in impaired water reabsorption via AQP2. This likely contributes to the declined concentrating ability of the kidneys and high circulating vasopressin levels in ADPKD patients.

Authors

Karla M. Márquez-Nogueras, Ryne M. Knutila, Virdjinija Vuchkovska, Charlie Yang, Patricia Outeda, Darren P. Wallace, Ivana Y. Kuo

×

PRDM16 acts as a therapeutic downstream target of TGF-β signaling in chronic kidney disease
Qian Yuan, … , Youhua Liu, Chun Zhang
Qian Yuan, … , Youhua Liu, Chun Zhang
Published August 4, 2025
Citation Information: JCI Insight. 2025. https://doi.org/10.1172/jci.insight.191458.
View: Text | PDF

PRDM16 acts as a therapeutic downstream target of TGF-β signaling in chronic kidney disease

  • Text
  • PDF
Abstract

Transforming growth factor beta (TGF-β) signaling is the master modulator of renal fibrosis. However, targeting drugs have failed to prevent the progression of chronic kidney disease (CKD) in clinical trials due to the extensive biological regulation of TGF-β signaling. It is necessary to investigate the precise downstream mechanisms of TGF-β signaling that regulate renal fibrosis. In this study, we found that PR-domain containing 16 (PRDM16) expression in human renal tubular epithelial cells was markedly reduced by TGF-β. Mechanistically, activated Smad3 induced by TGF-β interacted with the cofactor H-Ras and bound to the promoter of PRDM16, downregulating its transcription. Tubular-specific knockout of PRDM16 promoted renal fibrosis in models of unilateral ureteral occlusion (UUO) and unilateral ischemia-reperfusion injury (UIRI) by exacerbating mitochondrial dysfunction. In vitro, PRDM16 blocked TGF-β-induced mitochondrial injury and lipid deposition by upregulating Peroxisome Proliferator-Activated Receptor Gamma Coactivator-1α (PGC-1α). Delivery of the exogenous PRDM16 gene preserved renal function and ameliorated the progression of renal fibrosis by protecting mitochondrial function. We report PRDM16 as a novel downstream target of TGF-β signaling that attenuates renal fibrosis by safeguarding tubular mitochondrial function.

Authors

Qian Yuan, Ben Tang, Yuting Zhu, Chao Wan, Yaru Xie, Yajuan Xie, Cheng Wan, Hua Su, Youhua Liu, Chun Zhang

×

Low‐intensity pulsed ultrasound stimulation to treat renal fibrosis through inhibiting tubular IL-1R
Zhimin Huang, … , Xiangqing Kong, Huijuan Mao
Zhimin Huang, … , Xiangqing Kong, Huijuan Mao
Published July 29, 2025
Citation Information: JCI Insight. 2025. https://doi.org/10.1172/jci.insight.186892.
View: Text | PDF
Article has an altmetric score of 1

Low‐intensity pulsed ultrasound stimulation to treat renal fibrosis through inhibiting tubular IL-1R

  • Text
  • PDF
Abstract

Low-intensity pulsed ultrasound stimulation (LIPUS) has become increasingly appreciated for its therapeutic effect on kidney diseases. However, its role and biological mechanism in treating chronic kidney disease (CKD) remain poorly defined. Here, we revealed that LIPUS was applied in a safe range with an intensity of 25-315 mW/cm2. Daily LIPUS at an intensity of 315 mW/cm2 ameliorated ischemia-reperfusion (IR)-induced tubular injury and renal fibrosis, accompanied by the remarkable downregulation of IL-1R. Transcriptome sequencing showed that LIPUS significantly down-regulated IL-1R and its downstream genes in IL-1β-stimulated IR-injured mice. LIPUS effectively reversed IL-1β-induced tubular injury and reduced the production of profibrotic cytokines by down-regulating IL-1R in vivo and in vitro. Renal proximal tubule-specific Il1r1 knockout mice exhibited milder renal tubular injury and fibrosis after IR injury. However, LIPUS did not ameliorate IR injury in proximal tubule-specific Il1r1 knockout mice. Collectively, daily LIPUS at an intensity of 315 mW/cm2 relieves IR-induced tubular injury and fibrosis, potentially through down-regulating tubular IL-1R.

Authors

Zhimin Huang, Jiaxin Dong, Ziqi Fu, Li Li, Simeng liu, Lin Wu, Honglei Guo, Ao Bian, Kang Liu, Wei Sun, Changying Xing, Steven D. Crowley, Jiafa Ren, Xiangqing Kong, Huijuan Mao

×

NSD1-916aa encoded by CircNSD1 contributes to AKI-to-CKD transition through inducing ferroptosis in tubular epithelial cells
Li Gao, … , Ling Jiang, Yonggui Wu
Li Gao, … , Ling Jiang, Yonggui Wu
Published July 15, 2025
Citation Information: JCI Insight. 2025. https://doi.org/10.1172/jci.insight.189130.
View: Text | PDF

NSD1-916aa encoded by CircNSD1 contributes to AKI-to-CKD transition through inducing ferroptosis in tubular epithelial cells

  • Text
  • PDF
Abstract

Acute kidney injury (AKI) is characterized by a rapid decline in renal function. In severe or recurrent cases, AKI can progress to chronic kidney disease (CKD), marked by renal inflammation and fibrosis. Despite the severity of these outcomes, early-stage diagnostic tools and pharmacological interventions for AKI-to-CKD progression remain limited. In this study, we examined circular RNA (circRNA) expression profiles in mouse renal cortex tissues 14 days post-ischemia/reperfusion (I/R) injury using circRNA sequencing. The renal biopsy samples of patients after AKI exhibited reduced CircNSD1 expression, which was inversely associated with inflammation and fibrosis. Overexpression of CircNSD1 attenuated ferroptosis in vivo and in vitro, while slowing AKI-to-CKD progression. Mechanistically, CircNSD1 downregulated ACSL4 and SlC39A14 expression through histone H3 lysine 36 (H3K36) methylation, a critical pathway regulating ferroptosis after AKI or hypoxia/reoxygenation (H/R) injury. Furthermore, we identified that CircNSD1 encoded a NSD1-916aa peptide, which may functionally contribute to its observed effect. Collectively, these findings demonstrated that CircNSD1 may serve as a diagnostic and therapeutic target for early detection of AKI-to-CKD transition.

Authors

Li Gao, Junsheng Zhang, Chaoyi Chen, Sai Zhu, Xianglong Wei, Guiqin Tang, Sheng Wang, Yukai Wang, Xinran Liu, Ling Jiang, Yonggui Wu

×

Cullin-3 regulates the renal baroreceptor machinery that controls renin gene expression
Daria Golosova, … , Pablo Nakagawa, Curt D. Sigmund
Daria Golosova, … , Pablo Nakagawa, Curt D. Sigmund
Published July 8, 2025
Citation Information: JCI Insight. 2025. https://doi.org/10.1172/jci.insight.194075.
View: Text | PDF
Article has an altmetric score of 4

Cullin-3 regulates the renal baroreceptor machinery that controls renin gene expression

  • Text
  • PDF
Abstract

Mutations in Cullin-3 (CUL3) cause hypertension (HTN). We examined the role of smooth muscle cell (SMC) CUL3 in the regulation of renin gene expression. Mice with SMC-specific CUL3 deletion (S-CUL3KO) developed severe HTN with paradoxically preserved levels of plasma angiotensin peptides and renal renin expression. Cre-recombinase was active in JG cells resulting in decreased CUL3 expression. We evaluated components of the renin cell baroreceptor and revealed preserved lamin A/C but decreased integrin β1 expression in S-CUL3KO. We hypothesized that Rab proteins are involved in integrin β1 downregulation. Silencing either Rab21 or Rab5 in CUL3-deficient HEK293 cells increased integrin β1 protein. Co-immunoprecipitation revealed a direct interaction between Rab5 and CUL3. CUL3-deficiency increased Rab5 suggesting it is regulated by a CUL3-mediated mechanism and that CUL3-deficiency results in loss of Rab protein turnover leading to enhanced integrin β1 internalization. We conclude that the loss of integrin β1 from juxtaglomerular cells impairs the mechanosensory function of the renin cell baroreceptor, which underlies the persistent renin expression observed in hypertensive S-CUL3KO mice. These findings provide insights into the molecular mechanisms of HTN, revealing that dysregulation of Rab proteins and integrin β1 in the kidney due to CUL3-deficiency contributes to the development of HTN.

Authors

Daria Golosova, Gaurav Kumar, Ko-Ting Lu, Patricia C. Muskus Veitia, Ana Hantke Guixa, Kelsey K. Wackman, Eva M. Fekete, Daniel T. Brozoski, Justin L. Grobe, Maria Luisa S. Sequeira-Lopez, R. Ariel Gomez, Pablo Nakagawa, Curt D. Sigmund

×

The distal nephron biomarkers associate with diabetic kidney disease progression
Christina L. Tamargo, … , Joseph V. Bonventre, Chirag R. Parikh
Christina L. Tamargo, … , Joseph V. Bonventre, Chirag R. Parikh
Published June 23, 2025
Citation Information: JCI Insight. 2025;10(12):e186836. https://doi.org/10.1172/jci.insight.186836.
View: Text | PDF
Article has an altmetric score of 8

The distal nephron biomarkers associate with diabetic kidney disease progression

  • Text
  • PDF
Abstract

BACKGROUND While urinary biomarkers show promise in predicting diabetic kidney disease (DKD) progression, distal tubular markers remain understudied. We investigated the association of distal tubule markers, epidermal growth factor (EGF) and uromodulin (UMOD), with DKD progression in the Veterans Affairs Diabetes in Nephropathy (VA NEPHRON-D) clinical trial.METHODS. We used Cox regression models to evaluate the association between each biomarker and DKD progression and the relationship between change over time in biomarker and DKD progression. We used mixed models to investigate biomarker levels at baseline, 12 months, and over time and their relationships with longitudinal eGFR change.RESULTS. Participants (n = 1,116) had type 2 diabetes, urine albumin-to-creatinine ratio (UACR) ≥ 300 mg/g, and eGFR 30–89.9 mL/min/1.73 m2. Mean age was 65 years, mean eGFR was 56 (SD 19) mL/min/1.73 m2, and median UACR was 840 (IQR 424–1,780) mg/g. One hundred forty-four participants (13%) had DKD progression over a median follow-up of 2.2 (1.3–3.1) years. Higher baseline EGF and UMOD were independently associated with a lower risk of DKD progression (adjusted HR 0.68, 95% CI 0.47, 0.99 and 0.85, [0.75, 0.98] per 2-fold higher concentration of EGF and UMOD, respectively). Serial biomarker measurements were performed at baseline and 12 months, and a slower decline in biomarkers was associated with a lower risk of DKD progression when adjusted for baseline biomarker levels.CONCLUSION. Urinary EGF and UMOD may serve as valuable prognostic biomarkers in DKD.TRIAL REGISTRATION. ClinicalTrials.gov NCT00555217.FUNDING. NIH U01DK102730, U01DK103225, K23 DK118198, R01DK137087, U01DK103225, R37DK039773, U01DK114866, U01DK106962, U01DK129984, and R01DK093770; National Institute of Diabetes and Digestive and Kidney Diseases contract U01DK106965.

Authors

Christina L. Tamargo, Steven G. Coca, Heather Thiessen Philbrook, David G. Hu, Joachim H. Ix, Michael G. Shlipak, Linda F. Fried, Orlando M. Gutierrez, Sushrut S. Waikar, Sarah J. Schrauben, Jeffrey R. Schelling, Peter Ganz, Paul L. Kimmel, Jason H. Greenberg, Rajat Deo, Ayumi Takakura, Ramachandran S. Vasan, Joseph V. Bonventre, Chirag R. Parikh

×
  • ← Previous
  • 1
  • 2
  • 3
  • …
  • 26
  • 27
  • Next →

No posts were found with this tag.

Advertisement

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

Sign up for email alerts

Posted by 2 X users
Referenced by 1 Bluesky users
See more details
Posted by 3 X users
See more details
Referenced by 1 Bluesky users
See more details
Posted by 3 X users
See more details
Posted by 10 X users
On 1 Facebook pages
Referenced by 2 Bluesky users
3 readers on Mendeley
See more details
Posted by 5 X users
Referenced by 1 Bluesky users
See more details