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

In-Press Preview

  • 2,281 Articles
  • 0 Posts
  • ← Previous
  • 1
  • 2
  • 3
  • …
  • 228
  • 229
  • Next →
Small molecule correlates of infection precede infection diagnosis in breast implant reconstruction patients
John A. Wildenthal, Margaret A. Olsen, Hung D. Tran, John I. Robinson, Terence M. Myckatyn, David K. Warren, Keith E. Brandt, Marissa M. Tenenbaum, Joani M Christensen, Thomas H. Tung, Justin M. Sacks, Rachel A. Anolik, Katelin B. Nickel, Hideji Fujiwara, Peter J. Mucha, Jeffrey P. Henderson
John A. Wildenthal, Margaret A. Olsen, Hung D. Tran, John I. Robinson, Terence M. Myckatyn, David K. Warren, Keith E. Brandt, Marissa M. Tenenbaum, Joani M Christensen, Thomas H. Tung, Justin M. Sacks, Rachel A. Anolik, Katelin B. Nickel, Hideji Fujiwara, Peter J. Mucha, Jeffrey P. Henderson
View: Text | PDF

Small molecule correlates of infection precede infection diagnosis in breast implant reconstruction patients

  • Text
  • PDF
Abstract

BACKGROUND. Infection is an important complication of implanted devices and prosthetics. Identifying infections sufficiently early to salvage implants and avoid reconstructive failure is a persistent medical challenge. METHODS. Two female cohorts >21 years undergoing breast implant reconstruction were recruited. Seroma fluid (82 breasts, 70 patients) was collected upon implant removal for infectious or non-infectious causes. Post-implantation drain fluid (100 samples, 44 breasts, 32 patients) was collected at routine visits prior to implant removal. A liquid-chromatography/mass spectrometry-based metabolomic approach was used to identify infection correlates. RESULTS. In seroma fluid specimens, infection was associated with a diverse set of small molecules including acetylated polyamines, defensins, glucosyl-sphingosine, and several peptide-like features (all P<0.001, diagnostic areas under the receiver operating curve 0.82-0.93). Notably, a subset of these markers were significantly elevated (p<0.05) in post-implantation drain fluid before recorded infection symptoms and diagnosis. Pseudomonas aeruginosa and its specialized exometabolites in drain specimens were also associated with subsequent P. aeruginosa infections. CONCLUSION. Tissue fluid from infected patients has a distinctive metabolome reflecting human and bacterial physiologic processes that often precede clinical diagnoses. A diagnostic based on these findings has potential to improve patient outcomes through early recognition of infection. TRIAL REGISTRATION. Not applicable. FUNDING. Work was supported by U54CK000609 from the CDC and an unencumbered research gift to TMM from Sientra. Metabolomic approaches were supported by RO1DK125860 and RO1DK111930 to JPH. The contents are solely the responsibility of the authors and do not necessarily represent the official views of CDC.

Authors

John A. Wildenthal, Margaret A. Olsen, Hung D. Tran, John I. Robinson, Terence M. Myckatyn, David K. Warren, Keith E. Brandt, Marissa M. Tenenbaum, Joani M Christensen, Thomas H. Tung, Justin M. Sacks, Rachel A. Anolik, Katelin B. Nickel, Hideji Fujiwara, Peter J. Mucha, Jeffrey P. Henderson

×

Single cell characterization of the gastrointestinal HIV reservoir reveals heterogeneous cellular phenotypes
Jackson J. Peterson, Shipra Chandel, Katherine James, Elizabeth S. Bennett, Vincent Wu, Cory H. White, Brigitte Allard, Matthew Clohosey, Taylor Whitaker, Caroline Baker, Susan Pedersen, Anne F. Peery, Cynthia L. Gay, Michael R. Betts, David M. Margolis, Nancie M. Archin, Edward P. Browne
Jackson J. Peterson, Shipra Chandel, Katherine James, Elizabeth S. Bennett, Vincent Wu, Cory H. White, Brigitte Allard, Matthew Clohosey, Taylor Whitaker, Caroline Baker, Susan Pedersen, Anne F. Peery, Cynthia L. Gay, Michael R. Betts, David M. Margolis, Nancie M. Archin, Edward P. Browne
View: Text | PDF

Single cell characterization of the gastrointestinal HIV reservoir reveals heterogeneous cellular phenotypes

  • Text
  • PDF
Abstract

Human gastrointestinal (GI) tissues are a major site of HIV-1 viral persistence, but the nature of the GI reservoir remains poorly described. To characterize the GI HIV reservoir, we profiled cells from GI tissue and matched peripheral blood mononuclear cells from ten people with HIV on antiretroviral therapy using single cell RNA sequencing. We identified distinct compartment-specific patterns of gene expression, highlighting key differences between blood and colon CD4 T cell populations. vRNA+ cells from both blood and GI tissue were heterogeneous and found in multiple subtypes of CD4 T cells, although vRNA+ cells were particularly enriched in cells with Th17 or Treg17 phenotypes. Transcriptomic comparison of HIV vRNA+ and vRNA- T cells revealed 116 differentially expressed genes that were associated with HIV infection including ZBED2, MAF and IL17F. These data provide novel information regarding the GI-resident HIV reservoir and suggest that compartment-specific patterns of gene expression are associated with HIV infection.

Authors

Jackson J. Peterson, Shipra Chandel, Katherine James, Elizabeth S. Bennett, Vincent Wu, Cory H. White, Brigitte Allard, Matthew Clohosey, Taylor Whitaker, Caroline Baker, Susan Pedersen, Anne F. Peery, Cynthia L. Gay, Michael R. Betts, David M. Margolis, Nancie M. Archin, Edward P. Browne

×

Mismatch repair deficiency drives malignant progression and alters the tumor immune microenvironment in glioblastoma models
Montserrat Puigdelloses Vallcorba, Nishant Soni, Seung-Won Choi, Kavita Rawat, Tanvi Joshi, Sam Friedman, Alice Buonfiglioli, Angelo Angione, Zhihong Chen, Gonzalo Piñero, Gabrielle Price, Mehek Dedhia, Raina Roche, Emir Radkevich, Anne M. Bowcock, Deepti Bhatt, Winfried Edelmann, Robert M. Samstein, Timothy E. Richardson, Nadejda M. Tsankova, Alexander M. Tsankov, Ranjit S. Bindra, Raul Rabadan, Juan C. Vasquez, Dolores Hambardzumyan
Montserrat Puigdelloses Vallcorba, Nishant Soni, Seung-Won Choi, Kavita Rawat, Tanvi Joshi, Sam Friedman, Alice Buonfiglioli, Angelo Angione, Zhihong Chen, Gonzalo Piñero, Gabrielle Price, Mehek Dedhia, Raina Roche, Emir Radkevich, Anne M. Bowcock, Deepti Bhatt, Winfried Edelmann, Robert M. Samstein, Timothy E. Richardson, Nadejda M. Tsankova, Alexander M. Tsankov, Ranjit S. Bindra, Raul Rabadan, Juan C. Vasquez, Dolores Hambardzumyan
View: Text | PDF

Mismatch repair deficiency drives malignant progression and alters the tumor immune microenvironment in glioblastoma models

  • Text
  • PDF
Abstract

Mutations in DNA mismatch repair (MMR) pathway genes (MSH2, MSH6, MLH1, and PMS2) are linked to acquired resistance to temozolomide (TMZ) and high tumor mutation burden (TMB) in high-grade gliomas (HGG), including glioblastoma (GBM). However, the specific roles of individual MMR genes in the initiation, progression, TMB, microsatellite instability (MSI), and resistance to TMZ in glioma remain unclear. Here, we developed de novo mouse models of germline and somatic MMR-deficient (MMRd) HGG. Surprisingly, loss of Msh2 or Msh6 does not lead to high TMB, MSI, nor confer response to anti-PD-1 in GBM. Similarly, human GBM shows discordance between MMR gene mutations and TMB/MSI.Germline MMRd leads to promoted progression from low-grade to HGG and reduced survival compared to MMR-proficient (MMRp) tumor-bearing mice. This effect is not tumor cell intrinsic but is associated with MMRd in the tumor immune microenvironment, driving immunosuppressive myeloid programs, reduced lymphoid infiltration, and CD8+ T cell exhaustion. Both MMR-reduced (MMRr) and MMRd GBM are resistant to temozolomide (TMZ), unlike MMRp tumors. Our study shows that KL-50, a imidazotetrazine-based DNA targeting agent inducing MMR-independent cross-link–mediated cytotoxicity, was effective against germline and somatic MMRr/MMRd GBM, offering a potential therapy for TMZ-resistant HGG with MMR alterations.

Authors

Montserrat Puigdelloses Vallcorba, Nishant Soni, Seung-Won Choi, Kavita Rawat, Tanvi Joshi, Sam Friedman, Alice Buonfiglioli, Angelo Angione, Zhihong Chen, Gonzalo Piñero, Gabrielle Price, Mehek Dedhia, Raina Roche, Emir Radkevich, Anne M. Bowcock, Deepti Bhatt, Winfried Edelmann, Robert M. Samstein, Timothy E. Richardson, Nadejda M. Tsankova, Alexander M. Tsankov, Ranjit S. Bindra, Raul Rabadan, Juan C. Vasquez, Dolores Hambardzumyan

×

Hepatic steatosis in humans is associated with preserved glucagon action on amino acid metabolism
Hannah E. Christie, Sneha Mohan, Aoife M. Egan, Federica Boscolo, Chiara Dalla Man, Scott M. Thompson, Michael Jundt, Chad J. Fleming, James C. Andrews, Kent R. Bailey, Michael D. Jensen, K. Sree Nair, Adrian Vella
Hannah E. Christie, Sneha Mohan, Aoife M. Egan, Federica Boscolo, Chiara Dalla Man, Scott M. Thompson, Michael Jundt, Chad J. Fleming, James C. Andrews, Kent R. Bailey, Michael D. Jensen, K. Sree Nair, Adrian Vella
View: Text | PDF

Hepatic steatosis in humans is associated with preserved glucagon action on amino acid metabolism

  • Text
  • PDF
Abstract

BACKGROUND. Amino acid (AA) concentrations are increased in prediabetes and diabetes. Since AA stimulate glucagon secretion which should then increase hepatic AA catabolism, it has been hypothesized that hepatic resistance (associated with hepatic fat content) to glucagon’s actions on AA metabolism leads to hyperglucagonemia and hyperglycemia. METHODS. To test this hypothesis, we therefore studied lean and obese individuals, the latter group with and without hepatic steatosis as defined by Proton Density Fat Fraction (PDFF) > 5%. After an overnight fast, femoral vein, femoral artery, and hepatic vein catheters were placed. [3-3H] glucose and L-[1-13C,15N]-leucine were used to measure glucose turnover and leucine oxidation respectively. During a hyperglycemic clamp, an amino acid mixture was infused together with insulin and glucagon (1.5 ng/kg/min 0 – 120 min; 3.0 ng/kg/min 120 – 240 min). Tracer-based measurement of hepatic leucine oxidation in response to rising glucagon concentrations and splanchnic balance (measured using arterio-venous differences across the liver), of the other AA were the main outcomes measured. RESULTS. The presence of hepatic steatosis did not alter hepatic glucose metabolism and leucine oxidation in response to insulin and rising concentrations of glucagon. Splanchnic balance of a few amino acids, and related metabolites differed amongst the groups. However, across-group differences of AA splanchnic balance in response to glucagon were unaffected by the presence of hepatic steatosis. CONCLUSION. The action of glucagon on hepatic amino acid metabolism is unaffected by hepatic steatosis in humans. TRIAL REGISTRATION. This study was registered at Clinical Trials.Gov: NCT05500586. FUNDING. This work was funding by the NIH.

Authors

Hannah E. Christie, Sneha Mohan, Aoife M. Egan, Federica Boscolo, Chiara Dalla Man, Scott M. Thompson, Michael Jundt, Chad J. Fleming, James C. Andrews, Kent R. Bailey, Michael D. Jensen, K. Sree Nair, Adrian Vella

×

Mitochondrial Oxidants Promote Platelet Activation and Thrombotic Susceptibility in Prediabetes
Azaj Ahmed, Pooja Yadav, Melissa Jensen, Katharine Geasland, Jagadish S. Swamy, Douglas R. Spitz, E. Dale Abel, Diana Jalal, Sanjana Dayal
Azaj Ahmed, Pooja Yadav, Melissa Jensen, Katharine Geasland, Jagadish S. Swamy, Douglas R. Spitz, E. Dale Abel, Diana Jalal, Sanjana Dayal
View: Text | PDF

Mitochondrial Oxidants Promote Platelet Activation and Thrombotic Susceptibility in Prediabetes

  • Text
  • PDF
Abstract

Recent studies suggest that prediabetes is an independent risk factor for cardiovascular thrombotic events. However, the mechanisms that may promote platelet activation and thrombosis in prediabetes remain elusive. To determine mechanisms linking prediabetes and thrombosis as a function of age, we recruited prediabetic and normoglycemic Veterans in young and middle-age groups. Compared to normoglycemic subjects, platelets from those with prediabetes exhibited increased activation, mitochondrial-oxidant load, mitochondrial-membrane hyperpolarization, and greater thrombus formation ex vivo regardless of age. Preincubation of platelets with mitochondria targeted antioxidants such as superoxide dismutase (SOD) mimetic or Mito quinol (MitoQ), rescued this prothrombotic phenotype. These phenotypes were recapitulated in C57BL6/J mice exhibiting early onset of glucose intolerance when fed high fat (HF) diet for two weeks. Treatment of HF-fed mice with a SOD-mimetic or MitoQ, or genetic overexpression of catalase within mitochondria, not only lowered mitochondrial-oxidants, hyperpolarization, Ca2+ levels and platelet activation, but also protected against increased potential for carotid and pulmonary thrombosis. We also observed a bidirectional regulation of platelet activation by Ca2+ and mitochondrial oxidants. These findings support the idea that mitochondrial-oxidant dependent platelet activation induces a prothrombotic state in clinical prediabetes and preclinical models of short-term glucose intolerance and can be reversed by mitochondria-targeted antioxidants.

Authors

Azaj Ahmed, Pooja Yadav, Melissa Jensen, Katharine Geasland, Jagadish S. Swamy, Douglas R. Spitz, E. Dale Abel, Diana Jalal, Sanjana Dayal

×

PPP2R2A insufficiency enhances PD-L1 immune checkpoint blockade efficacy in lung cancer through cGAS-STING activation
Zhaojun Qiu, No-Joon Song, Anqi Li, Deepika Singh, Chandra B. Prasad, Chunhong Yan, David P. Carbone, Qi-en Wang, Xiaoli Zhang, Zihai Li, Junran Zhang
Zhaojun Qiu, No-Joon Song, Anqi Li, Deepika Singh, Chandra B. Prasad, Chunhong Yan, David P. Carbone, Qi-en Wang, Xiaoli Zhang, Zihai Li, Junran Zhang
View: Text | PDF

PPP2R2A insufficiency enhances PD-L1 immune checkpoint blockade efficacy in lung cancer through cGAS-STING activation

  • Text
  • PDF
Abstract

PP2A B55α, a regulatory subunit of protein phosphatase 2 (PP2A), is underexpressed in over 40% of non-small cell lung cancer (NSCLC) cases due to loss of heterozygosity of PPP2R2A, the gene encoding this protein. Given that low PPP2R2A expression correlates with poor prognosis, treating PPP2R2A-deficient NSCLC represents an unmet medical need. Here, we show that PPP2R2A knockdown or its heterozygosity (PPP2R2A+/–) increases cytosolic DNA, leading to cGAS-STING-type I interferon (IFN) pathway activation. PPP2R2A deficiency results in elevated expression of immune checkpoint protein PD-L1 via GSK-3β- and STING-dependent mechanisms. PPP2R2A+/– cancer cells have enhanced sensitivity to PD-L1 blockade in a mouse model of lung cancer due to modulation of the tumor immune microenvironment, resulting in increased NK cells and reduced infiltration and function of regulatory T cells (Tregs). Consequently, PD-L1 antibody treatment increases CD8+ T infiltration and activity, especially in tumors with PPP2R2A heterozygosity. Further, systemic or Treg-specific IFNAR1 blockade reduces the efficacy of PD-L1 blockade in PPP2R2A+/– tumors. Patients with NSCLC with a low PPP2R2A/PD-L1 ratio respond better to immune checkpoint blockade (ICB). These findings underscore the therapeutic potential of ICB in treating PPP2R2A-deficient NSCLC while suggesting that PPP2R2A deficiency could serve as a biomarker for guiding ICB-based therapies.

Authors

Zhaojun Qiu, No-Joon Song, Anqi Li, Deepika Singh, Chandra B. Prasad, Chunhong Yan, David P. Carbone, Qi-en Wang, Xiaoli Zhang, Zihai Li, Junran Zhang

×

MET alterations are enriched in lung adenocarcinoma brain metastases, defining a distinct biologic subtype
Timothy F. Burns, Sanja Dacic, Anish Chakka, Ethan Miller, Maria A. Velez, Ashwin Somasundaram, Saveri Bhattacharya, Autumn Gaither-Davis, Princey Devadassan, Jingxiao Jin, Vinod Kumar, Arjun Pennathur, Joanne Xiu, Matthew Oberley, Michael J. Glantz, Sonikpreet Aulakh, Uma R. Chandran, Riyue Bao, Curtis Tatsuoka, Laura P. Stabile
Timothy F. Burns, Sanja Dacic, Anish Chakka, Ethan Miller, Maria A. Velez, Ashwin Somasundaram, Saveri Bhattacharya, Autumn Gaither-Davis, Princey Devadassan, Jingxiao Jin, Vinod Kumar, Arjun Pennathur, Joanne Xiu, Matthew Oberley, Michael J. Glantz, Sonikpreet Aulakh, Uma R. Chandran, Riyue Bao, Curtis Tatsuoka, Laura P. Stabile
View: Text | PDF

MET alterations are enriched in lung adenocarcinoma brain metastases, defining a distinct biologic subtype

  • Text
  • PDF
Abstract

Non-small cell lung cancer (NSCLC) exhibits the highest rates of brain metastases (BM) among all solid tumors and presents a significant clinical challenge. The development of novel therapeutic strategies targeting BM is clearly needed. We identified a significant enrichment of MET amplification in lung adenocarcinoma (LUAD) BM compared to primary LUAD and extracranial metastases in oncogene driver-negative patients. Of note, MET amplified BM were responsive to MET inhibitors in vivo including models with acquired MET amplification at the time of metastasis. MET alterations (amplifications and/or mutations) were also more frequently detected in circulating tumor DNA from LUAD BM patients than in those without BM. MET altered BM also demonstrated unique genomic features compared to non-MET altered BM. Transcriptomic analyses revealed that in contrast to MET wildtype BM, MET amplified BM exhibited a more inflamed tumor microenvironment and displayed evidence of metabolic adaptation, particularly a reliance on glycolysis in contrast to oxidative phosphorylation in MET wildtype BM. Further, MET amplified BM demonstrated evidence of epithelial-mesenchymal transition signaling including increased expression of TWIST1. Patients with MET amplified BM had significantly shorter overall survival. These findings highlight MET amplification as a critical driver of LUAD BM, emphasizing its potential as a therapeutic target.

Authors

Timothy F. Burns, Sanja Dacic, Anish Chakka, Ethan Miller, Maria A. Velez, Ashwin Somasundaram, Saveri Bhattacharya, Autumn Gaither-Davis, Princey Devadassan, Jingxiao Jin, Vinod Kumar, Arjun Pennathur, Joanne Xiu, Matthew Oberley, Michael J. Glantz, Sonikpreet Aulakh, Uma R. Chandran, Riyue Bao, Curtis Tatsuoka, Laura P. Stabile

×

Patient-Derived Xenograft Models of Fanconi Anemia-Associated Head and Neck Cancer Identify Personalized Therapeutic Strategies
Jennifer Grandis, Hua Li, Benjamin A. Harrison, Andrew L.H. Webster, Joanna Pucilowska, Austin Nguyen, Jinho Lee, Gordon B. Mills, Jovanka Gencel-Augusto, Yan Zeng, Steven R. Long, Mi-Ok Kim, Rex H. Lee, David I. Kutler, Theresa Scognamiglio, Margaret Brandwein-Weber, Mark Urken, Inna Khodos, Elisa de Stanchina, Yu-Chien Lin, Frank X. Donovan, Settara C. Chandrasekharappa, Moonjung Jung, Mathijs A. Sanders, Agata Smogorzewska, Daniel E. Johnson
Jennifer Grandis, Hua Li, Benjamin A. Harrison, Andrew L.H. Webster, Joanna Pucilowska, Austin Nguyen, Jinho Lee, Gordon B. Mills, Jovanka Gencel-Augusto, Yan Zeng, Steven R. Long, Mi-Ok Kim, Rex H. Lee, David I. Kutler, Theresa Scognamiglio, Margaret Brandwein-Weber, Mark Urken, Inna Khodos, Elisa de Stanchina, Yu-Chien Lin, Frank X. Donovan, Settara C. Chandrasekharappa, Moonjung Jung, Mathijs A. Sanders, Agata Smogorzewska, Daniel E. Johnson
View: Text | PDF

Patient-Derived Xenograft Models of Fanconi Anemia-Associated Head and Neck Cancer Identify Personalized Therapeutic Strategies

  • Text
  • PDF
Abstract

Fanconi anemia (FA) confers a high risk (~700-fold increase) of solid tumor formation, most often head and neck squamous cell carcinoma (HNSCC). FA germline DNA repair defects preclude administration of most chemotherapies and prior hematopoietic stem cell transplantation limits use of immunotherapy. Thus, surgery and judicious delivery of radiation offer the only treatment options, with most patients succumbing to their cancers. A paucity of preclinical models has limited the development of new treatments. Here, we report, to our knowledge, the first patient-derived xenografts (PDXs) of FA-HNSCC and highlight the efficacy of FDA-approved EGFR targeted therapies in tumors with high EGFR/p-EGFR levels and the activity of the FDA-approved Bcl-2 inhibitor venetoclax in a FA-HNSCC PDX overexpressing Bcl-2. These findings support the development of precision medicine approaches for FA-HNSCC.

Authors

Jennifer Grandis, Hua Li, Benjamin A. Harrison, Andrew L.H. Webster, Joanna Pucilowska, Austin Nguyen, Jinho Lee, Gordon B. Mills, Jovanka Gencel-Augusto, Yan Zeng, Steven R. Long, Mi-Ok Kim, Rex H. Lee, David I. Kutler, Theresa Scognamiglio, Margaret Brandwein-Weber, Mark Urken, Inna Khodos, Elisa de Stanchina, Yu-Chien Lin, Frank X. Donovan, Settara C. Chandrasekharappa, Moonjung Jung, Mathijs A. Sanders, Agata Smogorzewska, Daniel E. Johnson

×

Genetic disruption of mitochondrial dynamics and stasis leads to liver injury and tumorigenesis
Xiaowen Ma, Xiaoli Wei, Mengwei Niu, Chen Zhang, Zheyun Peng, Wanqing Liu, Junrong Yan, Xiaoyang Su, Lichun Ma, Shaolei Lu, Wei Cui, Hiromi Sesaki, Wei-Xing Zong, Hong-Min Ni, Wen-Xing Ding
Xiaowen Ma, Xiaoli Wei, Mengwei Niu, Chen Zhang, Zheyun Peng, Wanqing Liu, Junrong Yan, Xiaoyang Su, Lichun Ma, Shaolei Lu, Wei Cui, Hiromi Sesaki, Wei-Xing Zong, Hong-Min Ni, Wen-Xing Ding
View: Text | PDF

Genetic disruption of mitochondrial dynamics and stasis leads to liver injury and tumorigenesis

  • Text
  • PDF
Abstract

Mitochondrial fission is mediated by dynamin-related protein 1 (gene name DNM1L) and fusion by mitofusins (MFN1 and MFN2) and optic atrophy 1 (OPA1). The role of mitochondrial dynamics in liver disease and cancer remains poorly understood. We used single, double, and triple liver-specific knockout (KO) mice lacking mitochondrial fission and fusion proteins, along with systematic analyses of mitochondrial morphology, untargeted metabolomics, RNA sequencing, hydrodynamic tail vein injection of oncogenes, and human hepatocellular carcinoma samples. Liver-specific Dnm1l KO (L-Dnm1l) mice showed increased ALT levels and hepatic fibrosis, with spontaneous liver tumors developing by 12 to 18 months of age. L-Mfn1 KO and L-Mfn2 KO mice showed no significant liver damage or tumor development, although a small percentage of double knockout (DKO) mice developed tumors. Triple knockout of Dnm1l, Mfn1, and Mfn2 (TKO) mice experienced significantly reduced liver injury and fibrosis, along with decreased spontaneous and oncogene-induced tumorigenesis. L-Dnm1l KO mice showed increased activation of the cGAS-STING-interferon pathway and pyrimidine metabolism, which were significantly normalized in TKO mice. Deletion of hepatic cGas reduced both basal and oncogene-induced liver injury and tumor development in L-Dnm1l KO mice. These findings indicate that mitochondrial dynamics are crucial for maintaining hepatic pyrimidine metabolism and regulating the cGAS-STING-mediated immune response to prevent liver tumorigenesis.

Authors

Xiaowen Ma, Xiaoli Wei, Mengwei Niu, Chen Zhang, Zheyun Peng, Wanqing Liu, Junrong Yan, Xiaoyang Su, Lichun Ma, Shaolei Lu, Wei Cui, Hiromi Sesaki, Wei-Xing Zong, Hong-Min Ni, Wen-Xing Ding

×

UTX coordinates TCF1 and STAT3 to control progenitor CD8+ T cell fate in autoimmune diabetes
Ho-Chung Chen, Madison F. Bang, Hsing-Hui Wang, Karl B. Shpargel, Lisa A. Kohn, David Sailer, Shile Zhang, Ethan C. McCarthy, Maryamsadat Seyedsadr, Satchel Stevens, Caitlyn L. H. Pham, Zikang Zhou, Xihui Yin, Nicole M. Wilkinson, Esther M. Peluso, Christian Bustillos, Jessica G. Ortega, Lixin Yang, Ashlyn A. Buzzelli, Reina C. Capati, Dennis J. Chia, Steven D. Mittelman, Christina M. Reh, Jason K. Whitmire, Melissa G. Lechner, Willy Hugo, Maureen A. Su
Ho-Chung Chen, Madison F. Bang, Hsing-Hui Wang, Karl B. Shpargel, Lisa A. Kohn, David Sailer, Shile Zhang, Ethan C. McCarthy, Maryamsadat Seyedsadr, Satchel Stevens, Caitlyn L. H. Pham, Zikang Zhou, Xihui Yin, Nicole M. Wilkinson, Esther M. Peluso, Christian Bustillos, Jessica G. Ortega, Lixin Yang, Ashlyn A. Buzzelli, Reina C. Capati, Dennis J. Chia, Steven D. Mittelman, Christina M. Reh, Jason K. Whitmire, Melissa G. Lechner, Willy Hugo, Maureen A. Su
View: Text | PDF

UTX coordinates TCF1 and STAT3 to control progenitor CD8+ T cell fate in autoimmune diabetes

  • Text
  • PDF
Abstract

Type 1 Diabetes Mellitus (T1D) is a chronic disease caused by an unremitting autoimmune attack on pancreatic beta cells. This autoimmune chronicity is mediated by stem-like progenitor CD8+ T cells that continually repopulate the pool of beta cell-specific cytolytic effectors. Factors governing the conversion of progenitors to effectors, however, remain unclear. T1D has been linked to a chromosomal region (Xp13-p11) that contains the epigenetic regulator UTX, which suggests a key role for UTX in T1D pathogenesis. Here, we show that T cell-specific UTX deletion in NOD mice protects against T1D development. In T cells of NOD mice and T1D patients, UTX ablation resulted in the accumulation of CD8+ progenitor cells with concomitant deficiency of effectors, suggesting a key role for UTX in poising progenitors for transition to effectors. Mechanistically, UTX’s role in T1D was independent of its inherent histone demethylase activity but instead relied on binding with transcription factors (TCF1 and STAT3) to co-regulate genes important in the maintenance and differentiation of progenitor CD8+ T cells. Together, these findings identify a critical role for UTX in T1D and the UTX:TCF1:STAT3 complex as a therapeutic target for terminating the long-lived autoimmune response.

Authors

Ho-Chung Chen, Madison F. Bang, Hsing-Hui Wang, Karl B. Shpargel, Lisa A. Kohn, David Sailer, Shile Zhang, Ethan C. McCarthy, Maryamsadat Seyedsadr, Satchel Stevens, Caitlyn L. H. Pham, Zikang Zhou, Xihui Yin, Nicole M. Wilkinson, Esther M. Peluso, Christian Bustillos, Jessica G. Ortega, Lixin Yang, Ashlyn A. Buzzelli, Reina C. Capati, Dennis J. Chia, Steven D. Mittelman, Christina M. Reh, Jason K. Whitmire, Melissa G. Lechner, Willy Hugo, Maureen A. Su

×
  • ← Previous
  • 1
  • 2
  • 3
  • …
  • 228
  • 229
  • 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