Go to JCI Insight
  • About
  • Editors
  • Consulting Editors
  • For authors
  • Publication ethics
  • Publication alerts by email
  • Advertising
  • Job board
  • Contact
  • Clinical Research and Public Health
  • Current issue
  • Past issues
  • By specialty
    • COVID-19
    • Cardiology
    • Gastroenterology
    • Immunology
    • Metabolism
    • Nephrology
    • Neuroscience
    • Oncology
    • Pulmonology
    • Vascular biology
    • All ...
  • Videos
    • Conversations with Giants in Medicine
    • Video Abstracts
  • Reviews
    • View all reviews ...
    • Clinical innovation and scientific progress in GLP-1 medicine (Nov 2025)
    • Pancreatic Cancer (Jul 2025)
    • Complement Biology and Therapeutics (May 2025)
    • Evolving insights into MASLD and MASH pathogenesis and treatment (Apr 2025)
    • Microbiome in Health and Disease (Feb 2025)
    • Substance Use Disorders (Oct 2024)
    • Clonal Hematopoiesis (Oct 2024)
    • View all review series ...
  • Viewpoint
  • Collections
    • In-Press Preview
    • Clinical Research and Public Health
    • Research Letters
    • Letters to the Editor
    • Editorials
    • Commentaries
    • Editor's notes
    • Reviews
    • Viewpoints
    • 100th anniversary
    • Top read articles

  • Current issue
  • Past issues
  • Specialties
  • Reviews
  • Review series
  • Conversations with Giants in Medicine
  • Video Abstracts
  • In-Press Preview
  • Clinical Research and Public Health
  • Research Letters
  • Letters to the Editor
  • Editorials
  • Commentaries
  • Editor's notes
  • Reviews
  • Viewpoints
  • 100th anniversary
  • Top read articles
  • About
  • Editors
  • Consulting Editors
  • For authors
  • Publication ethics
  • Publication alerts by email
  • Advertising
  • Job board
  • Contact

Research

  • 1,691 Articles
  • 0 Posts
  • ← Previous
  • 1
  • 2
  • 3
  • …
  • 169
  • 170
  • Next →
Limiting ER-associated degradation capacity triggers acute and chronic effects on insulin biosynthesis
Anoop Arunagiri, … , Ling Qi, Peter Arvan
Anoop Arunagiri, … , Ling Qi, Peter Arvan
Published November 18, 2025
Citation Information: J Clin Invest. 2025. https://doi.org/10.1172/JCI187341.
View: Text | PDF

Limiting ER-associated degradation capacity triggers acute and chronic effects on insulin biosynthesis

  • Text
  • PDF
Abstract

In pancreatic β-cells, misfolded proinsulin is a substrate for Endoplasmic Reticulum-Associated protein Degradation (ERAD) via HRD1/SEL1L. β-cell HRD1 activity is alternately reported to improve, or impair, insulin biogenesis. Further, while β-cell SEL1L deficiency causes HRD1 hypofunction and diminishes islet insulin content; reports conflict as to whether β-cell ERAD deficiency increases or decreases proinsulin levels. Here we’ve examined β-cell-specific Hrd1-KO mice (chronic deficiency), plus rodent (and human islet) β-cells treated acutely with HRD1 inhibitor. β-Hrd1-KO mice developed diabetes with decreased islet proinsulin yet a relative increase of misfolded proinsulin re-distributed to the ER; upregulated biochemical markers of β-cell ER stress and autophagy; electron microscopic evidence of ER enlargement and decreased insulin granule content; and increased glucagon-positive islet cells. Misfolded proinsulin was also increased in islets treated with inhibitors of lysosomal degradation. Preceding any loss of total proinsulin, acute HRD1 inhibition triggered increased nonnative proinsulin, increased phospho-eIF2ɑ with inhibited proinsulin synthesis, and increased LC3b-II (the abundance of which requires expression of SigmaR1). We posit a subset of proinsulin molecules undergoes HRD1-mediated disposal. When HRD1 is unavailable, misfolded proinsulin accumulates, accompanied by increased phospho-eIF2ɑ that limits further proinsulin synthesis, plus SigmaR1-dependent autophagy activation, ultimately lowering steady-state β-cell proinsulin (and insulin) levels — triggering diabetes.

Authors

Anoop Arunagiri, Leena Haataja, Maroof Alam, Noah F. Gleason, Emma Mastroianni, Chao-Yin Cheng, Sami Bazzi Onton, Jeffrey Knupp, Ibrahim Metawea, Anis Hassan, Dennis Larkin, Deyu Fang, Billy Tsai, Ling Qi, Peter Arvan

×

USP22 drives tumor immune evasion and checkpoint blockade resistance through EZH2-mediated epigenetic silencing of MHC-I
Kun Liu, … , Huiping Liu, Deyu Fang
Kun Liu, … , Huiping Liu, Deyu Fang
Published November 18, 2025
Citation Information: J Clin Invest. 2025. https://doi.org/10.1172/JCI193162.
View: Text | PDF

USP22 drives tumor immune evasion and checkpoint blockade resistance through EZH2-mediated epigenetic silencing of MHC-I

  • Text
  • PDF
Abstract

While immune checkpoint blockade (ICB) therapy has revolutionized the antitumor therapeutic landscape, it remains successful in only a small subset of cancer patients. Poor or loss of MHC-I expression has been implicated as a common mechanism of ICB resistance. Yet the molecular mechanisms underlying impaired MHC-I remain to be fully elucidated. Herein, we identified USP22 as a critical factor responsible for ICB resistance through suppressing MHC-I-mediated neoantigen presentation to CD8 T cells. Both genetic and pharmacologic USP22 inhibition increased immunogenicity and overcome anti-PD-1 immunotherapeutic resistance. At the molecular level, USP22 functions as a deubiquitinase for the methyltransferase EZH2, leading to transcriptional silencing of MHC-I gene expression. Targeted Usp22 inhibition resulted in increased tumoral MHC-I expression and consequently enhanced CD8 T cell killing, which was largely abrogated by Ezh2 reconstitution. Multiplexed immunofluorescence staining detected a strong reverse correlation between USP22 expression and both 2M expression and CD8+ T lymphocyte infiltration in solid tumors. Importantly, USP22 upregulation was associated with ICB immunotherapeutic resistance in patients with lung cancer. Collectively, this study highlights the role of USP22 as a diagnostic biomarker for ICB resistance and provides a potential therapeutic avenue to overcome the current ICB resistance through inhibition of USP22.

Authors

Kun Liu, Radhika Iyer, Yi Li, Jun Zhu, Zhaomeng Cai, Juncheng Wei, Yang Cheng, Amy Tang, Hai Wang, Qiong Gao, Nikita Lavanya Mani, Noah Marx, Beixue Gao, D. Martin Watterson, Seema A. Khan, William J. Gradishar, Huiping Liu, Deyu Fang

×

Retinol tracing within murine neural retina reveals cell type-specific retinol transport and distribution
Zachary J. Engfer, … , Philip D. Kiser, Krzysztof Palczewski
Zachary J. Engfer, … , Philip D. Kiser, Krzysztof Palczewski
Published November 18, 2025
Citation Information: J Clin Invest. 2025. https://doi.org/10.1172/JCI198648.
View: Text | PDF

Retinol tracing within murine neural retina reveals cell type-specific retinol transport and distribution

  • Text
  • PDF
Abstract

11-cis-Retinal is essential for light perception in mammalian photoreceptors (PRs), and aberrations in retinoid transformations cause severe retinal diseases. Understanding these processes is crucial for combating blinding diseases. The visual cycle, operating within PRs and the retinal pigment epithelium (RPE), regenerates 11-cis-retinal to sustain light sensitivity. Retinoids are also present in Müller glia (MG), hypothesized to supply 11-cis-retinol to cone PRs and retinal ganglion cells (RGCs). To trace retinoid movement through retinal cell types, we used cell-specific knock-in of lecithin:retinol acyltransferase (LRAT), which converts retinols into stable retinyl esters (REs). Ectopic LRAT expression in murine PRs, MG, and RGCs resulted in RE synthesis, with REs differing in abundance and isomeric composition across cell types under genetic and light-based perturbations. PR inner segments showed high 11-cis-RE content, suggesting a constant 11-cis-retinoid supply for pigment regeneration. In MG expressing LRAT, all-trans-REs were detected, contrasting with 11-cis-REs in PRs. The MG-specific LRAT phenotype mirrored the RE-rich human neural retina, suggesting human MG may utilize LRAT to maintain retinoid reservoirs. Our findings reveal tightly controlled retinoid flux throughout the mammalian retina supporting sustained vision, expanding understanding of the visual cycle to combat retinal diseases.

Authors

Zachary J. Engfer, Grazyna Palczewska, Samuel W. Du, Jianye Zhang, Zhiqian Dong, Carolline Rodrigues Menezes, Jun Wang, Jianming Shao, Budd A. Tucker, Robert F. Mullins, Rui Chen, Philip D. Kiser, Krzysztof Palczewski

×

EZH2 crosstalk with RNA methylation promotes prostate cancer progression through modulation of m6A autoregulation pathway
Yang Yi, … , Rendong Yang, Qi Cao
Yang Yi, … , Rendong Yang, Qi Cao
Published November 18, 2025
Citation Information: J Clin Invest. 2025. https://doi.org/10.1172/JCI195840.
View: Text | PDF

EZH2 crosstalk with RNA methylation promotes prostate cancer progression through modulation of m6A autoregulation pathway

  • Text
  • PDF
Abstract

N6-methyladenosine (m6A), the most predominant RNA modification in humans, participates in various fundamental and pathological bioprocesses. Dynamic manipulation of m6A deposition in the transcriptome is critical for cancer progression, while how this regulation is achieved remains understudied. Here, we report that in prostate cancer (PCa), Polycomb group (PcG) protein Enhancer of Zeste Homolog 2 (EZH2) exerts an additional function in m6A regulation via its enzymatic activity. Mechanistically, EZH2 methylates and stabilizes FOXA1 proteins from degradation, which in turn facilitates the transcription of m6A reader YTHDF1. Through activating an m6A autoregulation pathway, YTHDF1 enhances the translation of METTL14 and WTAP, two critical components of the m6A methyltransferase complex (MTC), and thereby upregulates the global m6A level in PCa cells. We further demonstrate that inhibiting the catalytic activity of EZH2 suppresses the translation process globally through targeting the YTHDF1-m6A axis. By disrupting both the expression and interaction of key m6A MTC subunits, combinational treatment of EZH2 degrader MS8815 and m6A inhibitor STM2457 mitigates prostate tumor growth synergistically. Together, our study decodes a previously hidden interrelationship between EZH2 and mRNA modification, which may be leveraged to advance the EZH2-targeting curative strategies in cancer.

Authors

Yang Yi, Joshua Fry, Chaehyun Yum, Rui Wang, Siqi Wu, Sharath Narayan, Qi Liu, Xingxing Zhang, Htoo Zarni Oo, Ning Xie, Yanqiang Li, Xinlei Gao, Xufen Yu, Xiaoping Hu, Qiaqia Li, Kemal Keseroglu, Ertuğrul M. Özbudak, Sarki A. Abdulkadir, Kaifu Chen, Jian Jin, Jonathan C. Zhao, Xuesen Dong, Daniel Arango, Rendong Yang, Qi Cao

×

Clonal hematopoiesis activates pro-calcific pathways in macrophages and promotes aortic valve stenosis
Wesley T. Abplanalp, … , Alexander G. Bick, Andreas M. Zeiher
Wesley T. Abplanalp, … , Alexander G. Bick, Andreas M. Zeiher
Published November 18, 2025
Citation Information: J Clin Invest. 2025. https://doi.org/10.1172/JCI171634.
View: Text | PDF

Clonal hematopoiesis activates pro-calcific pathways in macrophages and promotes aortic valve stenosis

  • Text
  • PDF
Abstract

Clonal hematopoiesis due to TET2-driver mutations (CH) is associated with coronary heart disease and worse prognosis among patients with aortic valve stenosis (AVS). However, it is unknown what role CH plays in the pathogenesis of AVS. In a meta-analysis of All Of Us, BioVU, and the UK Biobank, patients with CHIP exhibited an increased risk of AVS, with a higher risk among patients with TET2 or ASXL1 mutations. Single-cell RNA-sequencing of immune cells from AVS patients harboring TET2 CH-driver mutations revealed monocytes with heightened pro-inflammatory signatures and increased expression of pro-calcific paracrine signaling factors, most notably Oncostatin M (OSM). Secreted factors from TET2-silenced macrophages increased in vitro calcium deposition by mesenchymal cells, which was ablated by OSM silencing. Atheroprone Ldlr–/– mice receiving CH-mimicking Tet2–/– bone marrow transplants displayed greater calcium deposition in aortic valves. Together, these results demonstrate that monocytes with CH promote aortic valve calcification, and that patients with CH are at increased risk of AVS.

Authors

Wesley T. Abplanalp, Michael A. Raddatz, Bianca Schuhmacher, Silvia Mas-Peiro, María A. Zuriaga, Nuria Matesanz, José J. Fuster, Yash Pershad, Caitlyn Vlasschaert, Alexander J. Silver, Eric H. Farber-Eger, Yaomin Xu, Quinn S. Wells, Delara Shahidi, Sameen Fatima, Xiao Yang, Adwitiya A.P. Boruah, Akshay Ware, Maximilian Merten, Moritz von Scheidt, David John, Mariana Shumliakivska, Marion Muhly-Reinholz, Mariuca Vasa-Nicotera, Stefan Guenter, Michael R. Savona, Brian R. Lindman, Stefanie Dimmeler, Alexander G. Bick, Andreas M. Zeiher

×

Functional Characterization of SDHB Variants Clarifies Hereditary Pheochromocytoma and Paraganglioma Risk and Genotype–Phenotype Relationships
Sooyeon Lee, … , James M. Ford, Justin P. Annes
Sooyeon Lee, … , James M. Ford, Justin P. Annes
Published November 18, 2025
Citation Information: J Clin Invest. 2025. https://doi.org/10.1172/JCI198165.
View: Text | PDF

Functional Characterization of SDHB Variants Clarifies Hereditary Pheochromocytoma and Paraganglioma Risk and Genotype–Phenotype Relationships

  • Text
  • PDF
Abstract

Hereditary pheochromocytoma and paraganglioma (hPPGL) is caused by pathogenic mutations in succinate dehydrogenase (SDH) genes, commonly SDHB. However, over 80% of SDHB missense variants are classified as variants of uncertain significance (VUS), limiting clinical interpretation and diagnostic utility of germline testing. To provide functional evidence of SDHB allele pathogenicity or benignity, we developed a cellular complementation assay that quantifies intracellular succinate/fumarate ratios as a readout of SDH enzymatic activity. This assay reliably distinguished pathogenic from benign alleles with high fidelity, outperforming and complementing computational predictions. Functional assessment of patient-derived VUS alleles supported reclassification of 87% of tested variants and revealed that mutations in the iron–sulfur cluster domain were amorphic, while those at or beyond the C-terminal residue Tyr273 retained function. Variants associated with Leigh syndrome retained activity, consistent with their biallelic inheritance and distinct pathogenic mechanisms from SDHB-related tumorigenesis. Notably, hypomorphic pathogenic SDHB variants correlated with increased head and neck paraganglioma occurrence, revealing a genotype–phenotype relationship. Functional characterization of SDHB missense variants supports clinical classification, informs hPPGL risk stratification, and has immediate diagnostic impact.

Authors

Sooyeon Lee, Leor Needleman, Julie Park, Rebecca C. Schugar, Qianjin Guo, James M. Ford, Justin P. Annes

×

Curing autoimmune diabetes in mice with islet and hematopoietic cell transplantation after CD117 antibody-based conditioning
Preksha Bhagchandani, … , Judith A. Shizuru, Seung K. Kim
Preksha Bhagchandani, … , Judith A. Shizuru, Seung K. Kim
Published November 18, 2025
Citation Information: J Clin Invest. 2025. https://doi.org/10.1172/JCI190034.
View: Text | PDF

Curing autoimmune diabetes in mice with islet and hematopoietic cell transplantation after CD117 antibody-based conditioning

  • Text
  • PDF
Abstract

Mixed hematopoietic chimerism after allogeneic hematopoietic cell transplantation (HCT) promotes tolerance of transplanted donor-matched solid organs, corrects autoimmunity, and could transform therapeutic strategies for autoimmune type 1 diabetes (T1D). However, development of non-toxic bone marrow conditioning protocols is needed to expand clinical use. We developed a chemotherapy-free, non-myeloablative (NMA) conditioning regimen that achieves mixed chimerism and allograft tolerance across MHC barriers in NOD mice. We obtained durable mixed hematopoietic chimerism in prediabetic NOD mice using anti-c-Kit monoclonal antibody, T-cell depleting antibodies, JAK1/2 inhibition, and low-dose total body irradiation prior to transplantation of MHC-mismatched B6 hematopoietic cells, preventing diabetes in 100% of chimeric NOD:B6 mice. In overtly diabetic NOD mice, NMA conditioning followed by combined B6 HCT and islet transplantation durably corrected diabetes in 100% of chimeric mice without chronic immunosuppression or graft-versus-host disease (GVHD). Chimeric mice remained immunocompetent, as assessed by blood count recovery and rejection of 3rd party allogeneic islets. Adoptive transfer studies and analysis of autoreactive T cells confirmed correction of autoimmunity. Analysis of chimeric NOD mice revealed central thymic deletion and peripheral tolerance mechanisms. Thus, with NMA conditioning and cell transplantation, we achieved durable hematopoietic chimerism without GVHD, promoted islet allograft tolerance, and reversed established T1D.

Authors

Preksha Bhagchandani, Stephan A. Ramos, Bianca Rodriguez, Xueying Gu, Shiva Pathak, Yuqi Zhou, Yujin Moon, Nadia Nourin, Charles A. Chang, Jessica Poyser, Brenda J. Velasco, Weichen Zhao, Hye-Sook Kwon, Richard Rodriguez, Diego M. Burgos, Mario A. Miranda, Everett Meyer, Judith A. Shizuru, Seung K. Kim

×

TNF Superfamily Member 14 Drives Post-Influenza Depletion of Alveolar Macrophages Enabling Secondary Pneumococcal Pneumonia
Christina Malainou, … , Ulrich Matt, Susanne Herold
Christina Malainou, … , Ulrich Matt, Susanne Herold
Published November 18, 2025
Citation Information: J Clin Invest. 2025. https://doi.org/10.1172/JCI185390.
View: Text | PDF

TNF Superfamily Member 14 Drives Post-Influenza Depletion of Alveolar Macrophages Enabling Secondary Pneumococcal Pneumonia

  • Text
  • PDF
Abstract

Secondary bacterial infection, often caused by Streptococcus pneumoniae (Spn), is one of the most frequent and severe complications of influenza A virus (IAV)-induced pneumonia. Phenotyping of the pulmonary immune cell landscape after IAV infection revealed a substantial depletion of the tissue-resident alveolar macrophage (TR-AM) population at day 7, which was associated with increased susceptibility to Spn outgrowth. To elucidate the molecular mechanisms underlying TR-AM depletion, and to define putative targets for treatment, we combined single-cell transcriptomics and cell-specific PCR profiling in an unbiased manner, using in vivo models of IAV infection and IAV/Spn co-infection. The TNF superfamily 14 (TNFSF14) ligand-receptor axis was revealed as the driving force behind post-influenza TR-AM death during the early infection phase, enabling the transition to pneumococcal pneumonia, while intrapulmonary transfer of genetically modified TR-AMs and antibody-mediated neutralization of specific pathway components alleviated disease severity. With a mainly neutrophilic expression and a high abundance in the bronchoalveolar fluid (BALF) of patients with severe virus-induced ARDS, TNFSF14 emerged as a key determinant of virus-driven lung injury. Targeting the TNFSF14-mediated intercellular communication network in the virus-infected lung can, therefore, improve host defense, minimizing the risk of subsequent bacterial pneumonia, and ameliorating disease outcome.

Authors

Christina Malainou, Christin Peteranderl, Maximiliano Ruben Ferrero, Ana Ivonne Vazquez-Armendariz, Ioannis Alexopoulos, Katharina Franz, Klara Knippenberg, Julian Better, Mohammad Estiri, Cheng-Yu Wu, Hendrik Schultheis, Judith Bushe, Maria-Luisa del Rio, Jose Ignacio Rodriguez-Barbosa, Klaus Pfeffer, Stefan Günther, Mario Looso, Achim Dieter Gruber, István Vadász, Ulrich Matt, Susanne Herold

×

Impaired complement regulation drives chronic lung allograft dysfunction after lung transplantation
Hrishikesh S. Kulkarni, … , John R. Greenland, Andrew E. Gelman
Hrishikesh S. Kulkarni, … , John R. Greenland, Andrew E. Gelman
Published November 11, 2025
Citation Information: J Clin Invest. 2025. https://doi.org/10.1172/JCI188891.
View: Text | PDF

Impaired complement regulation drives chronic lung allograft dysfunction after lung transplantation

  • Text
  • PDF
Abstract

A greater understanding of chronic lung allograft dysfunction (CLAD) pathobiology, the primary cause of mortality after lung transplantation (LTx), is needed to improve outcomes. The complement system links innate to adaptive immune responses and is activated early post-lung transplantation to form the C3 convertase, a critical enzyme that cleaves the central complement component C3. We hypothesized that LTx recipients with a genetic predisposition to enhanced complement activation have worse CLAD-free survival mediated through increased adaptive alloimmunity. We interrogated a known functional C3 polymorphism (C3R102G) that increases complement activation through impaired C3 convertase inactivation in two independent LTx recipient cohorts. C3R102G, identified in at least one out of three LTx recipients, was associated with worse CLAD-free survival, particularly in the subset of recipients who developed donor-specific antibodies (DSAs). In a mouse orthotopic lung transplant model, impaired recipient complement regulation led to B cell-dependent CLAD pathology despite moderate differences in graft-infiltrating effector T cells. Dysregulated complement regulation promoted intragraft accumulation of memory B cells and antibody-secreting cells, leading to increased local and circulating DSA levels in mice. In summary, genetic predisposition to complement activation is associated with an increased humoral response and worse CLAD-free survival.

Authors

Hrishikesh S. Kulkarni, Laneshia K. Tague, Daniel R. Calabrese, Fuyi Liao, Zhiyi Liu, Lorena Garnica, Nishanth R. Shankar, Xiaobo Wu, Devesha H. Kulkarni, Aayusha Thapa, Dequan Zhou, Yan Tao, Victoria E. Davis, Cory T. Bernadt, Derek E. Byers, Catherine Chen, Howard J. Huang, Chad A. Witt, Ramsey R. Hachem, Daniel Kreisel, John P. Atkinson, John R. Greenland, Andrew E. Gelman

×

Lamin A/C regulates lipid metabolism and inflammation: insights from models of familial partial lipodystrophy 2
Jessica N. Maung, … , Elif A. Oral, Ormond A. MacDougald
Jessica N. Maung, … , Elif A. Oral, Ormond A. MacDougald
Published November 11, 2025
Citation Information: J Clin Invest. 2025. https://doi.org/10.1172/JCI198387.
View: Text | PDF

Lamin A/C regulates lipid metabolism and inflammation: insights from models of familial partial lipodystrophy 2

  • Text
  • PDF
Abstract

Familial partial lipodystrophy 2 (FPLD2) is a rare disease characterized by adipose tissue loss and redistribution, and metabolic dysfunction. FPLD2 is caused by pathogenic variants in the LMNA gene, encoding nuclear lamins A/C, structural proteins that control nuclear function and gene expression. However, the mechanisms driving adipocyte loss in FPLD2 remain poorly defined. In this study, we recruited eight families with developing or established FPLD2 and performed clinical, histological, and transcriptomic analyses of subcutaneous adipose tissue biopsies. Bulk and single-nuclei RNA-sequencing revealed suppression of lipid metabolism and mitochondrial pathways, alongside increased inflammation. These signatures were mirrored in tamoxifen-inducible adipocyte-specific Lmna knockout mice, in which lamin A/C-deficient adipocytes shrank and disappeared. Lmna-deficient fibroblasts shared similar gene expression changes, linked to altered chromatin accessibility, underscoring lamin A/C’s potential regulatory role in lipid metabolism and inflammatory programs. By directly comparing atrophic and hypertrophic adipose depots in FPLD2, and integrating human, mouse, and in vitro models, this study provides new insights into disease progression and potential therapeutic targets.

Authors

Jessica N. Maung, Rebecca L. Schill, Akira Nishii, Maria Foss de Freitas, Bonje N. Obua, Marcus Nygård, Maria D. Mendez-Casillas, Isabel D.K. Hermsmeyer, Donatella Gilio, Ozge Besci, Yang Chen, Brian Desrosiers, Rose E. Adler, Anabela D. Gomes, Merve Celik Guler, Hiroyuki Mori, Romina M. Uranga, Ziru Li, Hadla Hariri, Liping Zhang, Anderson de Paula Souza, Keegan S. Hoose, Kenneth T. Lewis, Taryn A. Hetrick, Paul Cederna, Carey N. Lumeng, Susanne Mandrup, Elif A. Oral, Ormond A. MacDougald

×
  • ← Previous
  • 1
  • 2
  • 3
  • …
  • 169
  • 170
  • Next →

No posts were found with this tag.

Advertisement

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

Sign up for email alerts