Pub Date : 2026-03-02Epub Date: 2025-12-12DOI: 10.1084/jem.20250777
Yongkang Yang, Qiaozhu Zuo, Vijay Ramu, Varen Talwar, Yajing Lyu, Chelsey Chen, Daiana Drehmer, Tina Yi-Ting Huang, Shaima Salman, Dominic Dordai, Yufeng Wang, Emmanuel Datan, Elizabeth E Wicks, Gregg L Semenza
Triple-negative breast cancer (TNBC) is the most aggressive breast cancer subtype with the highest rates of recurrence, metastasis, and patient mortality due to the absence of effective therapies. Hypoxia-inducible factor 1 (HIF-1) regulates the expression of thousands of RNAs in TNBC. Here, we demonstrate that transcription of the ISG20 gene, which encodes an RNA exonuclease, is activated by HIF-1 in TNBC cells. ISG20-mediated degradation of RHOBTB3 mRNA increases HIF-1α protein expression and activates NANOG signaling, which increases breast cancer stem cell specification and lung metastasis. ISG20 also degrades STAT1 and IRF1 mRNAs, leading to decreased expression of CXCL10 and impaired recruitment of CD8+ T cells and natural killer cells, thereby promoting breast cancer immune evasion. Silencing ISG20 increases the sensitivity of mouse TNBC cells to anti-PD1 antibody immune checkpoint blockade. Our data suggest that targeting ISG20, in combination with immunotherapy, could be an effective therapeutic strategy for TNBC.
{"title":"HIF-1-mediated ISG20 expression promotes breast cancer stemness and immune evasion.","authors":"Yongkang Yang, Qiaozhu Zuo, Vijay Ramu, Varen Talwar, Yajing Lyu, Chelsey Chen, Daiana Drehmer, Tina Yi-Ting Huang, Shaima Salman, Dominic Dordai, Yufeng Wang, Emmanuel Datan, Elizabeth E Wicks, Gregg L Semenza","doi":"10.1084/jem.20250777","DOIUrl":"https://doi.org/10.1084/jem.20250777","url":null,"abstract":"<p><p>Triple-negative breast cancer (TNBC) is the most aggressive breast cancer subtype with the highest rates of recurrence, metastasis, and patient mortality due to the absence of effective therapies. Hypoxia-inducible factor 1 (HIF-1) regulates the expression of thousands of RNAs in TNBC. Here, we demonstrate that transcription of the ISG20 gene, which encodes an RNA exonuclease, is activated by HIF-1 in TNBC cells. ISG20-mediated degradation of RHOBTB3 mRNA increases HIF-1α protein expression and activates NANOG signaling, which increases breast cancer stem cell specification and lung metastasis. ISG20 also degrades STAT1 and IRF1 mRNAs, leading to decreased expression of CXCL10 and impaired recruitment of CD8+ T cells and natural killer cells, thereby promoting breast cancer immune evasion. Silencing ISG20 increases the sensitivity of mouse TNBC cells to anti-PD1 antibody immune checkpoint blockade. Our data suggest that targeting ISG20, in combination with immunotherapy, could be an effective therapeutic strategy for TNBC.</p>","PeriodicalId":15760,"journal":{"name":"Journal of Experimental Medicine","volume":"223 3","pages":""},"PeriodicalIF":10.6,"publicationDate":"2026-03-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145743005","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-03-02Epub Date: 2025-12-05DOI: 10.1084/jem.20251760
Adrian Gervais, Astrid Marchal, Soraya Boucherit, Anthony Abi Haidar, Lucy Bizien, Ahmet Yalcinkaya, Ella Sandström, Xiao-Fei Kong, Emmanuel Jacquemin, Olivier Bernard, Dominique Debray, Florence Lacaille, Philippe Ichai, Cigdem Arikan, Etienne Javouhey, Bertrand Roquelaure, Frédéric Gottrand, Francesca Trespidi, Veronica Codullo, Lorenzo Cavagna, Nicolas Schleinitz, Mohamed Bousfiha, Naima Amenzoui, Ahmed Aziz Bousfiha, Sofie E Jørgensen, Nanna Mørk, Trine H Mogensen, Paul Bastard, Anne Puel, Alessandro Borghesi, Jody A Rule, William M Lee, Nils Landegren, Aurélie Cobat, Jean-Laurent Casanova, Emmanuelle Jouanguy
Fulminant viral hepatitis (FVH) is a devastating condition caused by hepatotropic viruses such as hepatitis A virus (HAV), hepatitis B virus (HBV), and HSV-1/2. We studied 149 FVH patients (73 males and 76 females, aged 1-76) for blood autoantibodies (auto-Abs) neutralizing type I interferons (IFNs; IFN-α2, -β, -ω). Six of 16 (37.5%) HSV-triggered FVH patients carried such auto-Abs on admission, including three with a previously known autoimmune disease. These patients contrasted with 133 HAV- (n = 46) or HBV-triggered (n = 87) patients, none of whom had such detectable auto-Abs. Odds ratios for HSV-triggered FVH in individuals with auto-Abs ranged from 35.3 (95% CI: 13.0-96.2; P < 10-7) for those neutralizing only 100 pg/ml IFN-α/ω to 1,895 (CI: 448.5-8,002; P < 10-12) for those neutralizing both IFN-α and IFN-ω at 10 ng/ml. Over one third of HSV-triggered FVH cases in this international cohort were due to preexisting auto-Abs. This finding highlights auto-Abs against type I IFNs as a major determinant of HSV-FVH and paves the way for targeted preventive or therapeutic interventions.
{"title":"Autoantibodies neutralizing type I IFNs in patients with fulminant herpes simplex virus hepatitis.","authors":"Adrian Gervais, Astrid Marchal, Soraya Boucherit, Anthony Abi Haidar, Lucy Bizien, Ahmet Yalcinkaya, Ella Sandström, Xiao-Fei Kong, Emmanuel Jacquemin, Olivier Bernard, Dominique Debray, Florence Lacaille, Philippe Ichai, Cigdem Arikan, Etienne Javouhey, Bertrand Roquelaure, Frédéric Gottrand, Francesca Trespidi, Veronica Codullo, Lorenzo Cavagna, Nicolas Schleinitz, Mohamed Bousfiha, Naima Amenzoui, Ahmed Aziz Bousfiha, Sofie E Jørgensen, Nanna Mørk, Trine H Mogensen, Paul Bastard, Anne Puel, Alessandro Borghesi, Jody A Rule, William M Lee, Nils Landegren, Aurélie Cobat, Jean-Laurent Casanova, Emmanuelle Jouanguy","doi":"10.1084/jem.20251760","DOIUrl":"10.1084/jem.20251760","url":null,"abstract":"<p><p>Fulminant viral hepatitis (FVH) is a devastating condition caused by hepatotropic viruses such as hepatitis A virus (HAV), hepatitis B virus (HBV), and HSV-1/2. We studied 149 FVH patients (73 males and 76 females, aged 1-76) for blood autoantibodies (auto-Abs) neutralizing type I interferons (IFNs; IFN-α2, -β, -ω). Six of 16 (37.5%) HSV-triggered FVH patients carried such auto-Abs on admission, including three with a previously known autoimmune disease. These patients contrasted with 133 HAV- (n = 46) or HBV-triggered (n = 87) patients, none of whom had such detectable auto-Abs. Odds ratios for HSV-triggered FVH in individuals with auto-Abs ranged from 35.3 (95% CI: 13.0-96.2; P < 10-7) for those neutralizing only 100 pg/ml IFN-α/ω to 1,895 (CI: 448.5-8,002; P < 10-12) for those neutralizing both IFN-α and IFN-ω at 10 ng/ml. Over one third of HSV-triggered FVH cases in this international cohort were due to preexisting auto-Abs. This finding highlights auto-Abs against type I IFNs as a major determinant of HSV-FVH and paves the way for targeted preventive or therapeutic interventions.</p>","PeriodicalId":15760,"journal":{"name":"Journal of Experimental Medicine","volume":"223 3","pages":""},"PeriodicalIF":10.6,"publicationDate":"2026-03-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12679989/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145677780","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-03-02Epub Date: 2025-12-05DOI: 10.1084/jem.20251962
Qian Zhang, Taylor S Conrad, Marcela Moncada-Velez, Kaijun Jiang, Anastasija Cupic, Jonathan Eaton, Kimberley Hutchinson, Adrian Gervais, Ruyue Chen, Anne Puel, Paul Bastard, Aurelie Cobat, Theresa Sokol, Ryan A Langlois, Lisa Miorin, Adolfo García-Sastre, John A Vanchiere, Jean-Laurent Casanova
Avian influenza A virus (IAV) H5N1 is an emerging threat of human pandemic. We describe a 71-year-old man who died of H5N1 pneumonia in Louisiana and whose blood contained autoantibodies neutralizing type I IFNs (AAN-I-IFNs), including the 12 IFN-α subtypes (1-10 ng/ml) and IFN-ω (100 pg/ml). Causality between these AAN-I-IFN and lethal outcome of avian influenza in this patient is based on (1) our previous report that AA-I-IFN underlie about 5% of cases of critical pneumonia triggered by seasonal influenza viruses in three cohorts, (2) the rarity of this combination of AAN-I-FNs in individuals over 70 years old (<1%), and (3) the rarity of lethal avian influenza among infected individuals (<1%). AAN-I-IFNs underlie a growing number of severe viral diseases, from arboviral encephalitis to viral pneumonia, particularly in the elderly. This case suggests they can also underlie life-threatening avian H5N1 influenza. The presence of AAN-I-IFN may facilitate infection, replication, and adaptation of zoonotic IAVs to humans and, therefore, human-to-human transmission.
{"title":"Autoantibodies neutralizing type I IFNs in a fatal case of H5N1 avian influenza.","authors":"Qian Zhang, Taylor S Conrad, Marcela Moncada-Velez, Kaijun Jiang, Anastasija Cupic, Jonathan Eaton, Kimberley Hutchinson, Adrian Gervais, Ruyue Chen, Anne Puel, Paul Bastard, Aurelie Cobat, Theresa Sokol, Ryan A Langlois, Lisa Miorin, Adolfo García-Sastre, John A Vanchiere, Jean-Laurent Casanova","doi":"10.1084/jem.20251962","DOIUrl":"10.1084/jem.20251962","url":null,"abstract":"<p><p>Avian influenza A virus (IAV) H5N1 is an emerging threat of human pandemic. We describe a 71-year-old man who died of H5N1 pneumonia in Louisiana and whose blood contained autoantibodies neutralizing type I IFNs (AAN-I-IFNs), including the 12 IFN-α subtypes (1-10 ng/ml) and IFN-ω (100 pg/ml). Causality between these AAN-I-IFN and lethal outcome of avian influenza in this patient is based on (1) our previous report that AA-I-IFN underlie about 5% of cases of critical pneumonia triggered by seasonal influenza viruses in three cohorts, (2) the rarity of this combination of AAN-I-FNs in individuals over 70 years old (<1%), and (3) the rarity of lethal avian influenza among infected individuals (<1%). AAN-I-IFNs underlie a growing number of severe viral diseases, from arboviral encephalitis to viral pneumonia, particularly in the elderly. This case suggests they can also underlie life-threatening avian H5N1 influenza. The presence of AAN-I-IFN may facilitate infection, replication, and adaptation of zoonotic IAVs to humans and, therefore, human-to-human transmission.</p>","PeriodicalId":15760,"journal":{"name":"Journal of Experimental Medicine","volume":"223 3","pages":""},"PeriodicalIF":10.6,"publicationDate":"2026-03-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12679982/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145677797","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-03-02Epub Date: 2025-12-16DOI: 10.1084/jem.20241287
Gabrielle R LeBlanc, Adam L Sobel, Jonathan Melamed, Dominic Haas, Eduard Ansaldo, Aiko M Cirone, Elizabeth Murguia, Michael G Constantinides
Mucosal-associated invariant T (MAIT) cells are predominantly located in barrier tissues where they rapidly respond to pathogens and commensals by recognizing microbial derivatives of riboflavin synthesis. Early-life exposure to these metabolites imprints the abundance of MAIT cells within tissues, so we hypothesized that antibiotic use during this period may abrogate their development. We identified antibiotics that deplete riboflavin-synthesizing commensals and revealed an early period of susceptibility during which antibiotic administration impaired MAIT cell development. The reduction in MAIT cell abundance rendered mice more susceptible to pneumonia, while MAIT cell-deficient mice were unaffected by early-life antibiotics. Concomitant administration of a riboflavin-synthesizing commensal during antibiotic treatment was sufficient to restore MAIT cell development and immunity. Our work demonstrates that transient depletion of riboflavin-synthesizing commensals in early life can adversely affect responses to subsequent infections.
{"title":"Antibiotic use in early life impairs MAIT cell-mediated immunity in adulthood.","authors":"Gabrielle R LeBlanc, Adam L Sobel, Jonathan Melamed, Dominic Haas, Eduard Ansaldo, Aiko M Cirone, Elizabeth Murguia, Michael G Constantinides","doi":"10.1084/jem.20241287","DOIUrl":"10.1084/jem.20241287","url":null,"abstract":"<p><p>Mucosal-associated invariant T (MAIT) cells are predominantly located in barrier tissues where they rapidly respond to pathogens and commensals by recognizing microbial derivatives of riboflavin synthesis. Early-life exposure to these metabolites imprints the abundance of MAIT cells within tissues, so we hypothesized that antibiotic use during this period may abrogate their development. We identified antibiotics that deplete riboflavin-synthesizing commensals and revealed an early period of susceptibility during which antibiotic administration impaired MAIT cell development. The reduction in MAIT cell abundance rendered mice more susceptible to pneumonia, while MAIT cell-deficient mice were unaffected by early-life antibiotics. Concomitant administration of a riboflavin-synthesizing commensal during antibiotic treatment was sufficient to restore MAIT cell development and immunity. Our work demonstrates that transient depletion of riboflavin-synthesizing commensals in early life can adversely affect responses to subsequent infections.</p>","PeriodicalId":15760,"journal":{"name":"Journal of Experimental Medicine","volume":"223 3","pages":""},"PeriodicalIF":10.6,"publicationDate":"2026-03-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145762960","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-03-02Epub Date: 2025-12-26DOI: 10.1084/jem.20241136
Carlota Farré Díaz, Eleni Kabrani, Wiebke Winkler, Eric Blanc, Brigitte Wollert-Wulf, Claudia Salomon, F Thomas Wunderlich, Dieter Beule, Martin Janz, Klaus Rajewsky
Modeling complex (patho)physiological processes by sequential targeted mutagenesis in mice is limited by the lack of precision of cellular targeting and complex breeding strategies. We present a new Cre/DreERT2 dual-recombinase germinal center B cell (GCBC)-specific strain, with co-expression of the recombinases from a single allele. This enables highly efficient Cre-mediated FOXO1 knockout in GCBCs in vivo, followed by time-controlled, efficient Dre-mediated FOXO1 re-expression in the same cells, leading to functional rescue of GC compartmentalization and class switch recombination. The present approach can be easily adapted to other cellular contexts.
{"title":"FOXO1 re-expression with a dual-recombinase allele rescues class switching in germinal center B cells.","authors":"Carlota Farré Díaz, Eleni Kabrani, Wiebke Winkler, Eric Blanc, Brigitte Wollert-Wulf, Claudia Salomon, F Thomas Wunderlich, Dieter Beule, Martin Janz, Klaus Rajewsky","doi":"10.1084/jem.20241136","DOIUrl":"10.1084/jem.20241136","url":null,"abstract":"<p><p>Modeling complex (patho)physiological processes by sequential targeted mutagenesis in mice is limited by the lack of precision of cellular targeting and complex breeding strategies. We present a new Cre/DreERT2 dual-recombinase germinal center B cell (GCBC)-specific strain, with co-expression of the recombinases from a single allele. This enables highly efficient Cre-mediated FOXO1 knockout in GCBCs in vivo, followed by time-controlled, efficient Dre-mediated FOXO1 re-expression in the same cells, leading to functional rescue of GC compartmentalization and class switch recombination. The present approach can be easily adapted to other cellular contexts.</p>","PeriodicalId":15760,"journal":{"name":"Journal of Experimental Medicine","volume":"223 3","pages":""},"PeriodicalIF":10.6,"publicationDate":"2026-03-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12755864/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145878414","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-03-02Epub Date: 2025-12-26DOI: 10.1084/jem.20251584
Cintia Bittar, Ana Rafaela Teixeira, Thiago Y Oliveira, Gabriela S Silva Santos, Klara Lenart, Marcilio Jorge Fumagalli, Georg H J Weymar, Anna Kaczynska, Noemi L Linden, Isabella A T M Ferreira, Marina Caskey, R Brad Jones, Mila Jankovic, Michel C Nussenzweig
Antiretroviral therapy suppresses HIV-1 infection but fails to eliminate a reservoir of intact latent proviruses that reside primarily in CD4+ T cells. The lack of precise understanding of the latent compartment has made it challenging to develop curative strategies for HIV-1 infection. Here we report on the properties of CD4+ T cell clones carrying intact latent proviruses, expanded in vitro from single cells obtained from the reservoir of people living with HIV-1. The latent proviruses in the clones were integrated into ZNF genes, nongenic satellite, and centromeric regions, frequently associated with latency. Despite their descent from single cells, only a fraction of the cells (0.4-14%) expressed relatively low levels of HIV-1 that did not measurably alter host gene transcriptome. Latency-reversing agents (LRAs) variably increased expression, but the effects were modest and clone and LRA specific. The results suggest that pharmacologic and immunologic approaches to clear the reservoir should be optimized to accommodate intra- and inter-clonal diversity.
{"title":"Transcription of HIV-1 is heterogenous among authentic latent CD4+ T cell clones.","authors":"Cintia Bittar, Ana Rafaela Teixeira, Thiago Y Oliveira, Gabriela S Silva Santos, Klara Lenart, Marcilio Jorge Fumagalli, Georg H J Weymar, Anna Kaczynska, Noemi L Linden, Isabella A T M Ferreira, Marina Caskey, R Brad Jones, Mila Jankovic, Michel C Nussenzweig","doi":"10.1084/jem.20251584","DOIUrl":"https://doi.org/10.1084/jem.20251584","url":null,"abstract":"<p><p>Antiretroviral therapy suppresses HIV-1 infection but fails to eliminate a reservoir of intact latent proviruses that reside primarily in CD4+ T cells. The lack of precise understanding of the latent compartment has made it challenging to develop curative strategies for HIV-1 infection. Here we report on the properties of CD4+ T cell clones carrying intact latent proviruses, expanded in vitro from single cells obtained from the reservoir of people living with HIV-1. The latent proviruses in the clones were integrated into ZNF genes, nongenic satellite, and centromeric regions, frequently associated with latency. Despite their descent from single cells, only a fraction of the cells (0.4-14%) expressed relatively low levels of HIV-1 that did not measurably alter host gene transcriptome. Latency-reversing agents (LRAs) variably increased expression, but the effects were modest and clone and LRA specific. The results suggest that pharmacologic and immunologic approaches to clear the reservoir should be optimized to accommodate intra- and inter-clonal diversity.</p>","PeriodicalId":15760,"journal":{"name":"Journal of Experimental Medicine","volume":"223 3","pages":""},"PeriodicalIF":10.6,"publicationDate":"2026-03-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145878488","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-03-02Epub Date: 2025-12-11DOI: 10.1084/jem.20250471
Urs M Mörbe, Fredrik V Junghus, Grigorii Nos, Peter B Jørgensen, Melissa J Ensmenger, Venla A Väänänen, Mads D Wewer, Gorm R Madsen, Lene B Riis, Henrik L Jakobsen, Lars R Olsen, Søren Brunak, Ole H Nielsen, William W Agace
Gut-associated lymphoid tissues (GALT) represent major sites of adaptive immune priming in the intestine, yet our understanding of human GALT diversity and function remains limited. Here, we used single-cell RNA sequencing, flow cytometry, and confocal laser microscopy to map the fibroblast (FB) landscape of human GALT, including that of Peyer's patches (PP), mucosal isolated lymphoid follicles (M-ILF), and submucosal ILF (SM-ILF). We identify CD24 as a marker that distinguishes GALT from other intestinal FB and demonstrate that CD24+ FB consist of distinct subsets that locate within discrete niches. We show that the composition and transcriptional profile of M-ILF and SM-ILF FB differs with SM-ILF FB appearing more focused at providing T cell support. Finally, we find the transcription profile of PP T zone reticular cells to be altered in Crohn's disease and that cells with a GALT FB-like profile can be detected in other chronic inflammatory diseases. Collectively, our findings provide an important framework for understanding GALT diversity and function.
{"title":"Fibroblast diversity within human gut-associated lymphoid tissues.","authors":"Urs M Mörbe, Fredrik V Junghus, Grigorii Nos, Peter B Jørgensen, Melissa J Ensmenger, Venla A Väänänen, Mads D Wewer, Gorm R Madsen, Lene B Riis, Henrik L Jakobsen, Lars R Olsen, Søren Brunak, Ole H Nielsen, William W Agace","doi":"10.1084/jem.20250471","DOIUrl":"10.1084/jem.20250471","url":null,"abstract":"<p><p>Gut-associated lymphoid tissues (GALT) represent major sites of adaptive immune priming in the intestine, yet our understanding of human GALT diversity and function remains limited. Here, we used single-cell RNA sequencing, flow cytometry, and confocal laser microscopy to map the fibroblast (FB) landscape of human GALT, including that of Peyer's patches (PP), mucosal isolated lymphoid follicles (M-ILF), and submucosal ILF (SM-ILF). We identify CD24 as a marker that distinguishes GALT from other intestinal FB and demonstrate that CD24+ FB consist of distinct subsets that locate within discrete niches. We show that the composition and transcriptional profile of M-ILF and SM-ILF FB differs with SM-ILF FB appearing more focused at providing T cell support. Finally, we find the transcription profile of PP T zone reticular cells to be altered in Crohn's disease and that cells with a GALT FB-like profile can be detected in other chronic inflammatory diseases. Collectively, our findings provide an important framework for understanding GALT diversity and function.</p>","PeriodicalId":15760,"journal":{"name":"Journal of Experimental Medicine","volume":"223 3","pages":""},"PeriodicalIF":10.6,"publicationDate":"2026-03-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12697342/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145723646","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-03-02Epub Date: 2026-01-05DOI: 10.1084/jem.20251067
Ian M Mbano, Nuo Liu, Marc H Wadsworth, Mark J Chambers, Thabo Mpotje, Osaretin E Asowata, Sarah K Nyquist, Kievershen Nargan, Duran Ramsuran, Farina Karim, Travis K Hughes, Joshua D Bromley, Robert Krause, Threnesan Naidoo, Liku B Tezera, Michaela T Reichmann, Sharie Keanne Ganchua, Henrik N Kløverpris, Kaylesh J Dullabh, Rajhmun Madansein, Sergio Triana, Adrie J C Steyn, Bonnie Berger, Mohlopheni J Marakalala, Gabriele Pollara, Sarah M Fortune, JoAnne L Flynn, Paul T Elkington, Alex K Shalek, Alasdair Leslie
Tuberculosis (TB) typically causes lung destruction and fibrosis, leading to ∼1.3 million deaths annually. The cellular drivers of human TB immunopathology remain poorly defined. We performed single-cell RNA sequencing and spatial transcriptomics on lung tissues from TB-infected and TB-negative individuals, identifying 30 distinct immune, parenchymal, and stromal cell subsets. Several were linked to TB pathology and corroborated through immunohistochemistry, flow cytometry, and independent human datasets. Fibroblasts were identified as major drivers in both active TB granuloma and TB-diseased lung tissue. In particular, the MMP1+CXCL5+ fibroblast subset, expressing a myofibroblast-like gene signature, was associated with severe disease and higher bacterial burden in nonhuman primate granulomas. Network analyses revealed cross talk between MMP1+CXCL5+ fibroblasts and SPP1+ macrophages within the granuloma cuff, which has been reported in other disease contexts, and may play an important role in TB immunopathology. Our findings highlight previously unappreciated cell populations and potential targets for novel TB therapies.
{"title":"Single-cell and spatial profiling highlights TB-induced myofibroblasts as drivers of lung pathology.","authors":"Ian M Mbano, Nuo Liu, Marc H Wadsworth, Mark J Chambers, Thabo Mpotje, Osaretin E Asowata, Sarah K Nyquist, Kievershen Nargan, Duran Ramsuran, Farina Karim, Travis K Hughes, Joshua D Bromley, Robert Krause, Threnesan Naidoo, Liku B Tezera, Michaela T Reichmann, Sharie Keanne Ganchua, Henrik N Kløverpris, Kaylesh J Dullabh, Rajhmun Madansein, Sergio Triana, Adrie J C Steyn, Bonnie Berger, Mohlopheni J Marakalala, Gabriele Pollara, Sarah M Fortune, JoAnne L Flynn, Paul T Elkington, Alex K Shalek, Alasdair Leslie","doi":"10.1084/jem.20251067","DOIUrl":"10.1084/jem.20251067","url":null,"abstract":"<p><p>Tuberculosis (TB) typically causes lung destruction and fibrosis, leading to ∼1.3 million deaths annually. The cellular drivers of human TB immunopathology remain poorly defined. We performed single-cell RNA sequencing and spatial transcriptomics on lung tissues from TB-infected and TB-negative individuals, identifying 30 distinct immune, parenchymal, and stromal cell subsets. Several were linked to TB pathology and corroborated through immunohistochemistry, flow cytometry, and independent human datasets. Fibroblasts were identified as major drivers in both active TB granuloma and TB-diseased lung tissue. In particular, the MMP1+CXCL5+ fibroblast subset, expressing a myofibroblast-like gene signature, was associated with severe disease and higher bacterial burden in nonhuman primate granulomas. Network analyses revealed cross talk between MMP1+CXCL5+ fibroblasts and SPP1+ macrophages within the granuloma cuff, which has been reported in other disease contexts, and may play an important role in TB immunopathology. Our findings highlight previously unappreciated cell populations and potential targets for novel TB therapies.</p>","PeriodicalId":15760,"journal":{"name":"Journal of Experimental Medicine","volume":"223 3","pages":""},"PeriodicalIF":10.6,"publicationDate":"2026-03-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12767585/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145900568","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-03-02Epub Date: 2025-12-17DOI: 10.1084/jem.20250535
Liling Xu, Maria Bottermann, Paula M Villavicencio, John Warner, Stephanie R Weldon, Zhenfei Xie, Andrew Filby, Xiaotie Liu, Ian G Ganley, Alison E Ringel, Usha Nair, Facundo D Batista
Little is known about the role of autophagy in the human humoral immune system. Here, we found that in B cells, genetic ablation of FIP200, a mammalian metabolic sensor that regulates autophagy in response to a range of stimuli, led to diminished humoral immune responses in mice. FIP200-deficient B cells displayed decreased differentiation into plasma cells, as well as mitochondrial dysfunction, alterations in heme biosynthesis, and significant cell death. Notably, the addition of heme was sufficient to rescue plasma cell differentiation of FIP200-deficient B cells. Thus, FIP200 determines B cell fates by controlling mitophagy and metabolic reprogramming.
{"title":"FIP200 regulates plasma B cell differentiation via mitochondrial and heme homeostasis.","authors":"Liling Xu, Maria Bottermann, Paula M Villavicencio, John Warner, Stephanie R Weldon, Zhenfei Xie, Andrew Filby, Xiaotie Liu, Ian G Ganley, Alison E Ringel, Usha Nair, Facundo D Batista","doi":"10.1084/jem.20250535","DOIUrl":"10.1084/jem.20250535","url":null,"abstract":"<p><p>Little is known about the role of autophagy in the human humoral immune system. Here, we found that in B cells, genetic ablation of FIP200, a mammalian metabolic sensor that regulates autophagy in response to a range of stimuli, led to diminished humoral immune responses in mice. FIP200-deficient B cells displayed decreased differentiation into plasma cells, as well as mitochondrial dysfunction, alterations in heme biosynthesis, and significant cell death. Notably, the addition of heme was sufficient to rescue plasma cell differentiation of FIP200-deficient B cells. Thus, FIP200 determines B cell fates by controlling mitophagy and metabolic reprogramming.</p>","PeriodicalId":15760,"journal":{"name":"Journal of Experimental Medicine","volume":"223 3","pages":""},"PeriodicalIF":10.6,"publicationDate":"2026-03-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12710606/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145768170","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-03-02Epub Date: 2025-12-09DOI: 10.1084/jem.20250607
Paulina D Horton, Alina Syed, Michelle Winkler, Abishek B Vaidya, Michael Rariden, Neha Arora, Yong Zhou, Michihiro Kobayashi, Momoko Yoshimoto, Hyun Jung Lee, Hyun-Eui Kim, John P Hagan, Catherine Denicourt, Travis I Moore, Pamela L Wenzel
Mechanical force generated by blood flow stimulates emergence of the first hematopoietic stem cells (HSCs) that populate the blood system. Force drives the transition of HSC precursors from an endothelial to hematopoietic identity, yet the molecular regulation of this fate switch remains poorly understood. We report that shear stress triggers adaptation in mitochondrial composition, ultrastructure, and function, which are essential for hematopoietic fate and engraftment potential. Shear stress remodels mitochondria in hemogenic endothelium by promoting mitochondrial gene transcription and protein synthesis. Laminar flow selectively initiates translation of 5' terminal polypyrimidine (5'TOP) motif-containing transcripts, which commonly encode ribosome and translation machinery. Flow-responsive metabolic reprogramming depends upon mechanistic target of rapamycin (mTOR) activation and is stymied when ribosome activity or mTOR is blocked. Conversely, chemical induction of mTOR mimics the effects of force on mitochondria and blood reconstituting potential and also partially rescues hematopoiesis in heartbeat mutants in utero. These findings identify mechanometabolism as a determinant of hematopoietic fate that could inform engineering of HSCs for disease modeling and treatment.
{"title":"Mechanometabolism instructs hematopoietic stem cell specification.","authors":"Paulina D Horton, Alina Syed, Michelle Winkler, Abishek B Vaidya, Michael Rariden, Neha Arora, Yong Zhou, Michihiro Kobayashi, Momoko Yoshimoto, Hyun Jung Lee, Hyun-Eui Kim, John P Hagan, Catherine Denicourt, Travis I Moore, Pamela L Wenzel","doi":"10.1084/jem.20250607","DOIUrl":"10.1084/jem.20250607","url":null,"abstract":"<p><p>Mechanical force generated by blood flow stimulates emergence of the first hematopoietic stem cells (HSCs) that populate the blood system. Force drives the transition of HSC precursors from an endothelial to hematopoietic identity, yet the molecular regulation of this fate switch remains poorly understood. We report that shear stress triggers adaptation in mitochondrial composition, ultrastructure, and function, which are essential for hematopoietic fate and engraftment potential. Shear stress remodels mitochondria in hemogenic endothelium by promoting mitochondrial gene transcription and protein synthesis. Laminar flow selectively initiates translation of 5' terminal polypyrimidine (5'TOP) motif-containing transcripts, which commonly encode ribosome and translation machinery. Flow-responsive metabolic reprogramming depends upon mechanistic target of rapamycin (mTOR) activation and is stymied when ribosome activity or mTOR is blocked. Conversely, chemical induction of mTOR mimics the effects of force on mitochondria and blood reconstituting potential and also partially rescues hematopoiesis in heartbeat mutants in utero. These findings identify mechanometabolism as a determinant of hematopoietic fate that could inform engineering of HSCs for disease modeling and treatment.</p>","PeriodicalId":15760,"journal":{"name":"Journal of Experimental Medicine","volume":"223 3","pages":""},"PeriodicalIF":10.6,"publicationDate":"2026-03-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12782086/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145708376","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}