Exosomes play a role in cell communication by transporting content between cells. Here, we tested whether renal podocyte-derived exosomes affect the injury of glomerular endothelial cells in lupus nephritis (LN). We found that exosomes containing high levels of high mobility group box 1 (HMGB1) were released from podocytes in patients with LN, BALB/c mice injected with pristane (which induces lupus-like disease in mice), and cultured human renal glomerular endothelial cells (HRGECs) treated with LN plasma. In vitro, GW4869 (an inhibitor of exosome biogenesis/release) or exosome removal alleviated the injury of HRGECs induced by LN plasma. Additionally, leptomycin B or knockdown of HMGB1 in podocyte-derived exosomes reduced endothelial cell injury and the expression of tripartite motif-containing 27 (TRIM27). Knockdown or overexpression of TRIM27 attenuated or promoted the damage of HRGECs treated with LN plasma. In vivo, knockdown of HMGB1 in podocytes ameliorated the injury of glomerular endothelial cells (GECs) in a mouse model of LN. Furthermore, the injection of podocyte-derived exosomes into mice caused GEC dysfunction. In conclusion, our study revealed that podocyte-derived exosomes may mediate the injury of glomerular endothelial cells seen in LN.
{"title":"HMGB1 encapsulated in podocyte-derived exosomes plays a central role in glomerular endothelial cell injury in lupus nephritis by regulating TRIM27 expression.","authors":"Jinxi Liu, Tongyu Zhao, Huixin Cui, Yuexin Tian, Xinyan Miao, Lingling Xing, Xiaorong Wang, Jie Huang, Qingjuan Liu, Wei Zhang, Ke Shi, Yunhe Liu, Baiyun Jia, Lihua Kang, Yu Tian, Weicheng Yuan, Shiwei He, Xiaojuan Feng, Shuxia Liu","doi":"10.1016/j.labinv.2025.104096","DOIUrl":"https://doi.org/10.1016/j.labinv.2025.104096","url":null,"abstract":"<p><p>Exosomes play a role in cell communication by transporting content between cells. Here, we tested whether renal podocyte-derived exosomes affect the injury of glomerular endothelial cells in lupus nephritis (LN). We found that exosomes containing high levels of high mobility group box 1 (HMGB1) were released from podocytes in patients with LN, BALB/c mice injected with pristane (which induces lupus-like disease in mice), and cultured human renal glomerular endothelial cells (HRGECs) treated with LN plasma. In vitro, GW4869 (an inhibitor of exosome biogenesis/release) or exosome removal alleviated the injury of HRGECs induced by LN plasma. Additionally, leptomycin B or knockdown of HMGB1 in podocyte-derived exosomes reduced endothelial cell injury and the expression of tripartite motif-containing 27 (TRIM27). Knockdown or overexpression of TRIM27 attenuated or promoted the damage of HRGECs treated with LN plasma. In vivo, knockdown of HMGB1 in podocytes ameliorated the injury of glomerular endothelial cells (GECs) in a mouse model of LN. Furthermore, the injection of podocyte-derived exosomes into mice caused GEC dysfunction. In conclusion, our study revealed that podocyte-derived exosomes may mediate the injury of glomerular endothelial cells seen in LN.</p>","PeriodicalId":17930,"journal":{"name":"Laboratory Investigation","volume":" ","pages":"104096"},"PeriodicalIF":5.1,"publicationDate":"2025-01-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143029128","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cardiovascular disease (CVD) is the leading cause of death worldwide and has been confirmed to be associated with a common oral bacterial infection-chronic apical periodontitis (CAP). However, the detailed mechanisms remain controversial. CAP can potentially alter systemic inflammation, lipid metabolism, and gut microbiota, all of which contribute to the progression of the aortic inflammatory response. This study aimed to explore the differential effects between E. faecalis and P. gingivalis-CAP on the aortic inflammatory response, which focused on changes in systemic inflammation, lipid metabolism, and gut microbiota, to explore potential mechanisms linking oral disease to CVD. Our result showed P. gingivalis-CAP could activate more serious aortic inflammatory cytokine mRNA expression (TNF-α, MCP-1, and ICAM-1) than E. faecalis-CAP by promoting higher serum inflammation (TNF-α, IL-6, IL-1α, and MCP-1) and lipid (LDL-C and TC) level. Simultaneously, there was no significant change in gut microbiota between them. Furthermore, all serum inflammatory cytokines showed substantial correlations with aortic inflammatory cytokine mRNA expression, and certain serum lipid indicators showed significant correlations, but only two gut microorganisms (Ruminococcaceae and Prevotellaceae) showed significant correlations. The combined results suggest that CAP might activate the aortic inflammatory response in association with changes in the three potential mechanisms. However, the promotion of gut microbiota might be relatively weak. Using experimental CAP induced by specific bacteria, in which bacteria are sequestered in the medullary cavity, avoids the direct influence of blood or intestinal pathways, and provides new perspectives for studying the mechanism of CVD associated with oral disease. Overall, these findings suggest that CAP may exacerbate systemic inflammation and serum lipid levels in patients with CVD, highlighting the importance of educating such patients on oral hygiene.
{"title":"Mono-species bacteria-induced chronic apical periodontitis triggers the aortic inflammatory response via modulation of systemic inflammation and lipid metabolism.","authors":"Huaxiang Lei, Shuai Chen, Xiaojing Huang, Dianfu Ma, Yufang Luo, Suli Xiao, Pingping Li, Guowu Gan, Zhiyu Cai","doi":"10.1016/j.labinv.2025.104095","DOIUrl":"https://doi.org/10.1016/j.labinv.2025.104095","url":null,"abstract":"<p><p>Cardiovascular disease (CVD) is the leading cause of death worldwide and has been confirmed to be associated with a common oral bacterial infection-chronic apical periodontitis (CAP). However, the detailed mechanisms remain controversial. CAP can potentially alter systemic inflammation, lipid metabolism, and gut microbiota, all of which contribute to the progression of the aortic inflammatory response. This study aimed to explore the differential effects between E. faecalis and P. gingivalis-CAP on the aortic inflammatory response, which focused on changes in systemic inflammation, lipid metabolism, and gut microbiota, to explore potential mechanisms linking oral disease to CVD. Our result showed P. gingivalis-CAP could activate more serious aortic inflammatory cytokine mRNA expression (TNF-α, MCP-1, and ICAM-1) than E. faecalis-CAP by promoting higher serum inflammation (TNF-α, IL-6, IL-1α, and MCP-1) and lipid (LDL-C and TC) level. Simultaneously, there was no significant change in gut microbiota between them. Furthermore, all serum inflammatory cytokines showed substantial correlations with aortic inflammatory cytokine mRNA expression, and certain serum lipid indicators showed significant correlations, but only two gut microorganisms (Ruminococcaceae and Prevotellaceae) showed significant correlations. The combined results suggest that CAP might activate the aortic inflammatory response in association with changes in the three potential mechanisms. However, the promotion of gut microbiota might be relatively weak. Using experimental CAP induced by specific bacteria, in which bacteria are sequestered in the medullary cavity, avoids the direct influence of blood or intestinal pathways, and provides new perspectives for studying the mechanism of CVD associated with oral disease. Overall, these findings suggest that CAP may exacerbate systemic inflammation and serum lipid levels in patients with CVD, highlighting the importance of educating such patients on oral hygiene.</p>","PeriodicalId":17930,"journal":{"name":"Laboratory Investigation","volume":" ","pages":"104095"},"PeriodicalIF":5.1,"publicationDate":"2025-01-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143007961","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-16DOI: 10.1016/j.labinv.2025.104094
Fanny Beltzung, Van Linh Le, Ioana Molnar, Erwan Boutault, Claude Darcha, François Le Loarer, Myriam Kossai, Olivier Saut, Julian Biau, Frédérique Penault-Llorca, Emmanuel Chautard
The tumor microenvironment (TME) plays a critical role in cancer progression and therapeutic responsiveness, with the tumor immune microenvironment (TIME) being a key modulator. In head and neck squamous cell carcinomas (HNSCC), immune cell infiltration significantly influences the response to radiotherapy (RT). A better understanding of the TIME in HNSCC could help identify patients most likely to benefit from combining RT with immunotherapy. Standardized, cost-effective methods for studying TIME in HNSCC are currently lacking. This study aims to leverage deep learning (DL) to quantify immune cell densities using immunohistochemistry (IHC) in untreated oropharyngeal squamous cell carcinoma (OPSCC) biopsies of patients scheduled for curative RT, and to assess their prognostic value. We analyzed 84 pre-treatment formalin-fixed paraffin-embedded (FFPE) tumor biopsies from OPSCC patients. Immunohistochemistry was performed for CD3, CD8, CD20, CD163, and FOXP3, and whole slide images (WSIs) were digitized for analysis using a U-Net-based DL model. Two quantification approaches were applied: a cell-counting method and an area-based method. These methods were applied to stained regions. The DL model achieved high accuracy in detecting stained cells across all biomarkers. Strong correlations were found between our DL pipeline, the HALO® Image Analysis Platform, and the open-source QuPath software for estimating immune cell densities. Our DL pipeline provided an accurate and reproducible approach for quantifying immune cells in OPSCC. The area-based method demonstrated superior prognostic value for recurrence-free survival (RFS), when compared to the cell-counting method. Elevated densities of CD3, CD8, CD20, and FOXP3 were associated with improved RFS, while CD163 showed no significant prognostic association. These results highlight the potential of DL in digital pathology for assessing TIME and predicting patient outcomes.
{"title":"Leveraging Deep Learning for Immune Cell Quantification and Prognostic Evaluation in Radiotherapy-Treated Oropharyngeal Squamous Cell Carcinomas.","authors":"Fanny Beltzung, Van Linh Le, Ioana Molnar, Erwan Boutault, Claude Darcha, François Le Loarer, Myriam Kossai, Olivier Saut, Julian Biau, Frédérique Penault-Llorca, Emmanuel Chautard","doi":"10.1016/j.labinv.2025.104094","DOIUrl":"https://doi.org/10.1016/j.labinv.2025.104094","url":null,"abstract":"<p><p>The tumor microenvironment (TME) plays a critical role in cancer progression and therapeutic responsiveness, with the tumor immune microenvironment (TIME) being a key modulator. In head and neck squamous cell carcinomas (HNSCC), immune cell infiltration significantly influences the response to radiotherapy (RT). A better understanding of the TIME in HNSCC could help identify patients most likely to benefit from combining RT with immunotherapy. Standardized, cost-effective methods for studying TIME in HNSCC are currently lacking. This study aims to leverage deep learning (DL) to quantify immune cell densities using immunohistochemistry (IHC) in untreated oropharyngeal squamous cell carcinoma (OPSCC) biopsies of patients scheduled for curative RT, and to assess their prognostic value. We analyzed 84 pre-treatment formalin-fixed paraffin-embedded (FFPE) tumor biopsies from OPSCC patients. Immunohistochemistry was performed for CD3, CD8, CD20, CD163, and FOXP3, and whole slide images (WSIs) were digitized for analysis using a U-Net-based DL model. Two quantification approaches were applied: a cell-counting method and an area-based method. These methods were applied to stained regions. The DL model achieved high accuracy in detecting stained cells across all biomarkers. Strong correlations were found between our DL pipeline, the HALO® Image Analysis Platform, and the open-source QuPath software for estimating immune cell densities. Our DL pipeline provided an accurate and reproducible approach for quantifying immune cells in OPSCC. The area-based method demonstrated superior prognostic value for recurrence-free survival (RFS), when compared to the cell-counting method. Elevated densities of CD3, CD8, CD20, and FOXP3 were associated with improved RFS, while CD163 showed no significant prognostic association. These results highlight the potential of DL in digital pathology for assessing TIME and predicting patient outcomes.</p>","PeriodicalId":17930,"journal":{"name":"Laboratory Investigation","volume":" ","pages":"104094"},"PeriodicalIF":5.1,"publicationDate":"2025-01-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143007959","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-16DOI: 10.1016/j.labinv.2025.104093
Klára Pavlíčková, Jan Hojný, Petr Waldauf, Marián Švajdler, Pavel Dundr, Pavel Fabian, Eva Krkavcová, Jiří Dvořák, Romana Michálková, Iva Staniczková Zambo, Nikola Hájková, Miroslava Flídrová, Jan Laco, Helena Hornychová, Patricie Delongová, Jozef Škarda, Jan Hrudka, Radoslav Matěj
Extrapulmonary small cell neuroendocrine carcinoma (EP-SCNC) is a rare malignancy with a poor prognosis. Most patients with EP-SCNC have metastatic disease upon presentation, and their average overall survival (OS) is less than 12 months. Our study aimed to conduct a complex analysis of EP-SCNC. One hundred and eighty-one EP-SCNC tissue samples were subjected to a complex analysis. One hundred and fifty-five tumors were pure EP-SCNC, while 26 were combined tumors. Immunohistochemistry for ASCL1, NEUROD1, YAP1, POU2F3, Rb1, p53, cyclin D1, p16, PTEN, DLL3, PD-L1, CD56, synaptophysin, chromogranin A, and INSM1 was performed, and 128 samples were analyzed molecularly using next generation sequencing (NGS), comprising DNA and RNA analysis. Detailed results on immunohistochemical and molecular analyses were provided for each primary origin of EP-SCNC separately. Median survival for the whole cohort of patients was 8.94 months. Patient age (≥ 70 years), tumor mutational burden (TMB) < 15, and TP53 and BRCA2 mutations were negative prognostic factors. High expression of ASCL-1 was associated with shorter OS, whereas high expression of YAP1 was associated with longer OS. Patients with genitourinary tumors had significantly better OS than those with gastrointestinal tract EP-SCNC tumors. Rb1 expression loss was detected more often in genitourinary tract SCNCs. In contrast, p16 overexpression was found more often in genitourinary tract SCNCs. POU2F3 expression was detected more often in combined tumors, whereas NEUROD1 was detected more often in pure EP-SCNC. Regarding "druggable markers," DLL3 was expressed in 66% of tumors and PD-L1 in 17.4%. Detailed analyses of different prognostic and predictive markers are needed to better understand EP-SCNC biology and create more personalized therapy to improve patient prognosis.
{"title":"Molecular and Immunohistochemical Classification of Extrapulmonary Small Cell Neuroendocrine Carcinomas: A Study of 181 Cases.","authors":"Klára Pavlíčková, Jan Hojný, Petr Waldauf, Marián Švajdler, Pavel Dundr, Pavel Fabian, Eva Krkavcová, Jiří Dvořák, Romana Michálková, Iva Staniczková Zambo, Nikola Hájková, Miroslava Flídrová, Jan Laco, Helena Hornychová, Patricie Delongová, Jozef Škarda, Jan Hrudka, Radoslav Matěj","doi":"10.1016/j.labinv.2025.104093","DOIUrl":"https://doi.org/10.1016/j.labinv.2025.104093","url":null,"abstract":"<p><p>Extrapulmonary small cell neuroendocrine carcinoma (EP-SCNC) is a rare malignancy with a poor prognosis. Most patients with EP-SCNC have metastatic disease upon presentation, and their average overall survival (OS) is less than 12 months. Our study aimed to conduct a complex analysis of EP-SCNC. One hundred and eighty-one EP-SCNC tissue samples were subjected to a complex analysis. One hundred and fifty-five tumors were pure EP-SCNC, while 26 were combined tumors. Immunohistochemistry for ASCL1, NEUROD1, YAP1, POU2F3, Rb1, p53, cyclin D1, p16, PTEN, DLL3, PD-L1, CD56, synaptophysin, chromogranin A, and INSM1 was performed, and 128 samples were analyzed molecularly using next generation sequencing (NGS), comprising DNA and RNA analysis. Detailed results on immunohistochemical and molecular analyses were provided for each primary origin of EP-SCNC separately. Median survival for the whole cohort of patients was 8.94 months. Patient age (≥ 70 years), tumor mutational burden (TMB) < 15, and TP53 and BRCA2 mutations were negative prognostic factors. High expression of ASCL-1 was associated with shorter OS, whereas high expression of YAP1 was associated with longer OS. Patients with genitourinary tumors had significantly better OS than those with gastrointestinal tract EP-SCNC tumors. Rb1 expression loss was detected more often in genitourinary tract SCNCs. In contrast, p16 overexpression was found more often in genitourinary tract SCNCs. POU2F3 expression was detected more often in combined tumors, whereas NEUROD1 was detected more often in pure EP-SCNC. Regarding \"druggable markers,\" DLL3 was expressed in 66% of tumors and PD-L1 in 17.4%. Detailed analyses of different prognostic and predictive markers are needed to better understand EP-SCNC biology and create more personalized therapy to improve patient prognosis.</p>","PeriodicalId":17930,"journal":{"name":"Laboratory Investigation","volume":" ","pages":"104093"},"PeriodicalIF":5.1,"publicationDate":"2025-01-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143007960","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-10DOI: 10.1016/j.labinv.2025.104091
Elahe Shenasa, Shelby Thornton, Dongxia Gao, Felix Kommoss, Torsten O Nielsen
Immunotherapy has emerged as a new treatment modality in some soft tissue sarcomas, particularly for tumors associated with tertiary lymphoid structures (TLS). These structures are functional lymphoid aggregates, and their presence is indicative of an active anticancer immune response in the tumor microenvironment. The assessment of TLS as a predictive biomarker at scale on patient specimens remains challenging. While tissue microarrays could facilitate this assessment, it is unclear whether small microarray cores can represent and identify associated TLS responses. We sought to use multiplex immunohistochemistry to identify key components of TLS: T cells, B cells, and dendritic cells. The multiplex panels (CD3, CD20, CD208 and PNAd) were applied onto 80 cases both on tissue microarrays and on their cognate available full-faced sections from epithelioid sarcoma and dedifferentiated/well-differentiated liposarcoma case series. Tissue microarrays were digitally scored for the number of immune cells using the HALO image analysis platform, and cognate full-faced sections were visually evaluated for the presence of TLS. An independent validation set of soft tissue sarcomas (N=49) was stained with the CD3, CD20, and CD208, and scored by QuPath. A Combined Immune Marker (defined as the presence of more than more than 24% CD3+T cell, or 0.51% CD20+B cell, or more than 0.14% CD208+mature dendritic cell on tissue microarray core) is highly specific (100%) and moderately sensitive (61%) to predict the existence of TLS on full-faced sections. The Combined Immune Marker showed a sensitivity of 25% and specificity of 91% on the validation set. The Combined Immune Marker assessed on tissue microarrays is highly specific to infer the presence of TLS present on cognate full-faced sections. Therefore, despite the small area sampled, tissue microarrays may be utilized to assess the clinical value of TLS on datasets where specificity is critical and large sample size can mitigate low to moderate sensitivity.
{"title":"Immune Biomarkers on Tissue Microarray Cores Support the Presence of Adjacent Tertiary Lymphoid Structures in Soft Tissue Sarcoma.","authors":"Elahe Shenasa, Shelby Thornton, Dongxia Gao, Felix Kommoss, Torsten O Nielsen","doi":"10.1016/j.labinv.2025.104091","DOIUrl":"https://doi.org/10.1016/j.labinv.2025.104091","url":null,"abstract":"<p><p>Immunotherapy has emerged as a new treatment modality in some soft tissue sarcomas, particularly for tumors associated with tertiary lymphoid structures (TLS). These structures are functional lymphoid aggregates, and their presence is indicative of an active anticancer immune response in the tumor microenvironment. The assessment of TLS as a predictive biomarker at scale on patient specimens remains challenging. While tissue microarrays could facilitate this assessment, it is unclear whether small microarray cores can represent and identify associated TLS responses. We sought to use multiplex immunohistochemistry to identify key components of TLS: T cells, B cells, and dendritic cells. The multiplex panels (CD3, CD20, CD208 and PNAd) were applied onto 80 cases both on tissue microarrays and on their cognate available full-faced sections from epithelioid sarcoma and dedifferentiated/well-differentiated liposarcoma case series. Tissue microarrays were digitally scored for the number of immune cells using the HALO image analysis platform, and cognate full-faced sections were visually evaluated for the presence of TLS. An independent validation set of soft tissue sarcomas (N=49) was stained with the CD3, CD20, and CD208, and scored by QuPath. A Combined Immune Marker (defined as the presence of more than more than 24% CD3+T cell, or 0.51% CD20+B cell, or more than 0.14% CD208+mature dendritic cell on tissue microarray core) is highly specific (100%) and moderately sensitive (61%) to predict the existence of TLS on full-faced sections. The Combined Immune Marker showed a sensitivity of 25% and specificity of 91% on the validation set. The Combined Immune Marker assessed on tissue microarrays is highly specific to infer the presence of TLS present on cognate full-faced sections. Therefore, despite the small area sampled, tissue microarrays may be utilized to assess the clinical value of TLS on datasets where specificity is critical and large sample size can mitigate low to moderate sensitivity.</p>","PeriodicalId":17930,"journal":{"name":"Laboratory Investigation","volume":" ","pages":"104091"},"PeriodicalIF":5.1,"publicationDate":"2025-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142971594","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01Epub Date: 2024-11-13DOI: 10.1016/j.labinv.2024.102186
Rick Ursem, Justus L Groen, Martijn J A Malessy, Inge Briaire-de Bruijn, Liam A McDonnell, Bram P A M Heijs, Judith V M G Bovee
Malignant peripheral nerve sheath tumors (MPNSTs) are aggressive sarcomas arising from peripheral nerves, accounting for 3% to 5% of soft tissue sarcomas. MPNSTs often recur locally, leading to poor survival. Achieving tumor-free surgical margins is essential to prevent recurrence, but current methods for determining tumor margins are limited, highlighting the need for improved biomarkers. In this study we investigated the degree to which MPNST extends into nerves adjacent to tumors. Alterations to the lipidome of MPNST and adjacent peripheral nerves were assessed using spatial lipidomics. Tissue samples from 5 patients with MPNST were analyzed, revealing alterations of the lipid profile extending into the peripheral nerves beyond what was expected based on macroscopic and histologic observations. Integration of spatial lipidomics and high-resolution accurate-mass profiling identified distinct lipid profiles associated with healthy nerves, connective tissue, and tumors. Notably, histologically normal nerves exhibited myelin degradation and infiltration of protumoral M2 macrophages, particularly near the tumor. Furthermore, aberrant osmium staining patterns and loss of H3K27me3 staining in the absence of atypia were observed in a case with tumor recurrence. This exploratory study thereby highlights the changes occurring in the nerves affected by MPNST beyond what is visible on hematoxylin and eosin staining and provides leads for further biomarker studies, including aberrant osmium staining, to assess resection margins in MPNST.
{"title":"Spatial Lipidomics Reveals Myelin Defects and Protumor Macrophage Infiltration in Malignant Peripheral Nerve Sheath Tumor Adjacent Nerves.","authors":"Rick Ursem, Justus L Groen, Martijn J A Malessy, Inge Briaire-de Bruijn, Liam A McDonnell, Bram P A M Heijs, Judith V M G Bovee","doi":"10.1016/j.labinv.2024.102186","DOIUrl":"10.1016/j.labinv.2024.102186","url":null,"abstract":"<p><p>Malignant peripheral nerve sheath tumors (MPNSTs) are aggressive sarcomas arising from peripheral nerves, accounting for 3% to 5% of soft tissue sarcomas. MPNSTs often recur locally, leading to poor survival. Achieving tumor-free surgical margins is essential to prevent recurrence, but current methods for determining tumor margins are limited, highlighting the need for improved biomarkers. In this study we investigated the degree to which MPNST extends into nerves adjacent to tumors. Alterations to the lipidome of MPNST and adjacent peripheral nerves were assessed using spatial lipidomics. Tissue samples from 5 patients with MPNST were analyzed, revealing alterations of the lipid profile extending into the peripheral nerves beyond what was expected based on macroscopic and histologic observations. Integration of spatial lipidomics and high-resolution accurate-mass profiling identified distinct lipid profiles associated with healthy nerves, connective tissue, and tumors. Notably, histologically normal nerves exhibited myelin degradation and infiltration of protumoral M2 macrophages, particularly near the tumor. Furthermore, aberrant osmium staining patterns and loss of H3K27me3 staining in the absence of atypia were observed in a case with tumor recurrence. This exploratory study thereby highlights the changes occurring in the nerves affected by MPNST beyond what is visible on hematoxylin and eosin staining and provides leads for further biomarker studies, including aberrant osmium staining, to assess resection margins in MPNST.</p>","PeriodicalId":17930,"journal":{"name":"Laboratory Investigation","volume":" ","pages":"102186"},"PeriodicalIF":5.1,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142623199","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01Epub Date: 2024-11-13DOI: 10.1016/j.labinv.2024.102185
Chi Sing Ng, Jilong Qin
About 20% of human cancers harbor mutations of genes encoding switch/sucrose nonfermentable (SWI/SNF) complex subunits. Deficiency of subunits of the complex is present in 10% of non-small-cell lung cancers (NSCLC; SMARCA4/SMARCA2 deficient), 100% thoracic SMARCA4/A2-deficient undifferentiated tumors (TSADUDT; SMARCA4/A2 deficient), malignant rhabdoid tumor, and atypical/teratoid tumor (SMARCB1-deficient), >90% of small cell carcinoma of the ovary, hypercalcemic type (SMARCA4/SMARCA2 deficient), frequently in undifferentiated/dedifferentiated endometrial carcinoma (SMARCA4, SMARCA2, SMARCB1, and ARID1A/B deficient), 100% SMARCA4 deficient undifferentiated uterine sarcoma (SMARCA4 deficient); and in various other tumors from multifarious anatomical sites. Silencing of SWI/SNF gene expression may be genomically or epigenetically driven, causing loss of tumor suppression function or facilitating other oncogenic events. The SWI/SNF-deficient tumors share the phenotype of poor or no differentiation, often with a variable component of rhabdoid tumor cells. They present at advanced stages with poor prognosis. Rhabdoid tumor cell phenotype is a useful feature to prompt investigation for this group of tumors. In the thoracic space, the overlap in morphology, immunophenotype, genetics, and epigenetics of SMARCA4/A2-deficient NSCLC and TSADUDT appears more significant. This raises a possible nosologic relationship between TSADUDT and SMARCA4/A2-deficient NSCLC. Increased understanding of the genetics, epigenetics, and mechanisms of oncogenesis in these poor prognostic tumors, which are often resistant to conventional treatment, opens a new horizon of therapy for the tumors.
{"title":"Switch/Sucrose Nonfermentable-Deficient Tumors-Morphology, Immunophenotype, Genetics, Epigenetics, Nosology, and Therapy.","authors":"Chi Sing Ng, Jilong Qin","doi":"10.1016/j.labinv.2024.102185","DOIUrl":"10.1016/j.labinv.2024.102185","url":null,"abstract":"<p><p>About 20% of human cancers harbor mutations of genes encoding switch/sucrose nonfermentable (SWI/SNF) complex subunits. Deficiency of subunits of the complex is present in 10% of non-small-cell lung cancers (NSCLC; SMARCA4/SMARCA2 deficient), 100% thoracic SMARCA4/A2-deficient undifferentiated tumors (TSADUDT; SMARCA4/A2 deficient), malignant rhabdoid tumor, and atypical/teratoid tumor (SMARCB1-deficient), >90% of small cell carcinoma of the ovary, hypercalcemic type (SMARCA4/SMARCA2 deficient), frequently in undifferentiated/dedifferentiated endometrial carcinoma (SMARCA4, SMARCA2, SMARCB1, and ARID1A/B deficient), 100% SMARCA4 deficient undifferentiated uterine sarcoma (SMARCA4 deficient); and in various other tumors from multifarious anatomical sites. Silencing of SWI/SNF gene expression may be genomically or epigenetically driven, causing loss of tumor suppression function or facilitating other oncogenic events. The SWI/SNF-deficient tumors share the phenotype of poor or no differentiation, often with a variable component of rhabdoid tumor cells. They present at advanced stages with poor prognosis. Rhabdoid tumor cell phenotype is a useful feature to prompt investigation for this group of tumors. In the thoracic space, the overlap in morphology, immunophenotype, genetics, and epigenetics of SMARCA4/A2-deficient NSCLC and TSADUDT appears more significant. This raises a possible nosologic relationship between TSADUDT and SMARCA4/A2-deficient NSCLC. Increased understanding of the genetics, epigenetics, and mechanisms of oncogenesis in these poor prognostic tumors, which are often resistant to conventional treatment, opens a new horizon of therapy for the tumors.</p>","PeriodicalId":17930,"journal":{"name":"Laboratory Investigation","volume":" ","pages":"102185"},"PeriodicalIF":5.1,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142623221","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-26DOI: 10.1016/j.labinv.2024.102221
Keita Kouzu, Hironori Tsujimoto, Ines P Nearchou, Takahiro Einama, Takanori Watanabe, Hiroyuki Horiguchi, Yoji Kishi, Hitoshi Tsuda, Hideki Ueno
Tumor cell nuclear size (NS) indicates malignant potential in breast cancer; however, its clinical significance in esophageal squamous cell carcinoma (ESCC) is unknown. Artificial intelligence (AI) can quantitatively evaluate histopathological findings. The aim was to measure NS in ESCC using AI and elucidate its clinical significance. We investigated the relationship between NS assessed by AI and prognosis in 138 patients with ESCC who underwent curative esophagectomy. Hematoxylin and eosin-stained slides from the deepest tumor sections were digitized. Using HALO-AI DenseNet v2, we created a deep-learning classifier that identified tumor cells with an NS area >20 μm2. Median NS was 40.14 μm2, which was used to divide patients into NS-high and NS-low groups (n = 69 per group). Five-year overall survival (OS) and relapse-free survival rates were significantly lower in the NS-high group (43.2% and 39.6%) than in the NS-low group (67.7% and 49.6%). Multivariate analysis showed that greater tumor depth and NS-high status (hazard ratio: 1.79; P = .032) were independent risk factors for OS. In 77 cases with neoadjuvant chemotherapy, increased tumor depth and NS-high status (hazard ratio: 1.99; P = .048) were independent prognostic factors for unfavorable OS. Compared with the NS-low group, the NS-high group had significantly higher anisokaryosis, higher Ki-67 expression as calculated by AI analysis of immunostaining, and higher NS heterogeneity as examined by equidividing the tumors into square tiles. In conclusion, NS assessed by AI is a simple and useful prognostic factor for ESCC.
{"title":"Prognostic Impact of Tumor Cell Nuclear Size Assessed by Artificial Intelligence in Esophageal Squamous Cell Carcinoma.","authors":"Keita Kouzu, Hironori Tsujimoto, Ines P Nearchou, Takahiro Einama, Takanori Watanabe, Hiroyuki Horiguchi, Yoji Kishi, Hitoshi Tsuda, Hideki Ueno","doi":"10.1016/j.labinv.2024.102221","DOIUrl":"10.1016/j.labinv.2024.102221","url":null,"abstract":"<p><p>Tumor cell nuclear size (NS) indicates malignant potential in breast cancer; however, its clinical significance in esophageal squamous cell carcinoma (ESCC) is unknown. Artificial intelligence (AI) can quantitatively evaluate histopathological findings. The aim was to measure NS in ESCC using AI and elucidate its clinical significance. We investigated the relationship between NS assessed by AI and prognosis in 138 patients with ESCC who underwent curative esophagectomy. Hematoxylin and eosin-stained slides from the deepest tumor sections were digitized. Using HALO-AI DenseNet v2, we created a deep-learning classifier that identified tumor cells with an NS area >20 μm<sup>2</sup>. Median NS was 40.14 μm<sup>2</sup>, which was used to divide patients into NS-high and NS-low groups (n = 69 per group). Five-year overall survival (OS) and relapse-free survival rates were significantly lower in the NS-high group (43.2% and 39.6%) than in the NS-low group (67.7% and 49.6%). Multivariate analysis showed that greater tumor depth and NS-high status (hazard ratio: 1.79; P = .032) were independent risk factors for OS. In 77 cases with neoadjuvant chemotherapy, increased tumor depth and NS-high status (hazard ratio: 1.99; P = .048) were independent prognostic factors for unfavorable OS. Compared with the NS-low group, the NS-high group had significantly higher anisokaryosis, higher Ki-67 expression as calculated by AI analysis of immunostaining, and higher NS heterogeneity as examined by equidividing the tumors into square tiles. In conclusion, NS assessed by AI is a simple and useful prognostic factor for ESCC.</p>","PeriodicalId":17930,"journal":{"name":"Laboratory Investigation","volume":" ","pages":"102221"},"PeriodicalIF":5.1,"publicationDate":"2024-12-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142895790","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-13DOI: 10.1016/j.labinv.2024.102211
Noelia Díaz-Morales, Sandra M Sancho-Martínez, Eva M Baranda-Alonso, Isabel Fuentes-Calvo, Rebeca S Sidhu-Muñoz, Nuria Martín-Fernández, Francisco J López-Hernández, Carlos Martínez-Salgado
Acute kidney frailty (AKF) is a condition of increased susceptibility to acute kidney injury (AKI), an abrupt impairment of renal excretory function potentially leading to severe complications. Prevention of AKI relies on the recognition of risk factors contributing to AKF. At the population level, dehydration constitutes a predisposing factor for AKI. However, renal frailty may be context-specific, with variations among patients in the types of damage and the distinct pathological mechanisms. In this regard, we studied the combined effect of dehydration with other factors on renal homeostasis, such as increasing age and hypertension. AKF status was studied in rats bearing risk factors individually and in combination and was evaluated as the level of AKI induced by a triggering dose of cisplatin, which is known to be mildly nephrotoxic for young, healthy rats. AKI was assessed through parameters of renal function (including creatinine, urea, creatinine clearance, proteinuria, and fractional excretion of sodium) and histopathology of renal tissue specimens. The hydration status was measured by bioelectric impedance and other techniques. Water deprivation induces a dehydration state characterized by reductions in body weight and urinary flow and increases in hematocrit and plasma and urine osmolality. Bioelectric impedance showed a net loss of body water after water deprivation with no relevant changes in body mass distribution. Dehydration is not sufficient to predispose young control rats to intrinsic AKI. However, the combination of dehydration with advanced age or hypertension induces AKF evidenced by a magnified response of renal dysfunction (reduced filtration and tubular function) and tubular necrosis caused by low-dose cisplatin treatment. This study highlights the relevance of addressing AKF as a premorbid condition providing prophylactic opportunities and shows that dehydration differentially predisposes to prerenal and intrinsic AKI.
{"title":"Age and Hypertension Synergize With Dehydration to Cause Renal Frailty in Rats and Predispose Them to Intrinsic Acute Kidney Injury.","authors":"Noelia Díaz-Morales, Sandra M Sancho-Martínez, Eva M Baranda-Alonso, Isabel Fuentes-Calvo, Rebeca S Sidhu-Muñoz, Nuria Martín-Fernández, Francisco J López-Hernández, Carlos Martínez-Salgado","doi":"10.1016/j.labinv.2024.102211","DOIUrl":"10.1016/j.labinv.2024.102211","url":null,"abstract":"<p><p>Acute kidney frailty (AKF) is a condition of increased susceptibility to acute kidney injury (AKI), an abrupt impairment of renal excretory function potentially leading to severe complications. Prevention of AKI relies on the recognition of risk factors contributing to AKF. At the population level, dehydration constitutes a predisposing factor for AKI. However, renal frailty may be context-specific, with variations among patients in the types of damage and the distinct pathological mechanisms. In this regard, we studied the combined effect of dehydration with other factors on renal homeostasis, such as increasing age and hypertension. AKF status was studied in rats bearing risk factors individually and in combination and was evaluated as the level of AKI induced by a triggering dose of cisplatin, which is known to be mildly nephrotoxic for young, healthy rats. AKI was assessed through parameters of renal function (including creatinine, urea, creatinine clearance, proteinuria, and fractional excretion of sodium) and histopathology of renal tissue specimens. The hydration status was measured by bioelectric impedance and other techniques. Water deprivation induces a dehydration state characterized by reductions in body weight and urinary flow and increases in hematocrit and plasma and urine osmolality. Bioelectric impedance showed a net loss of body water after water deprivation with no relevant changes in body mass distribution. Dehydration is not sufficient to predispose young control rats to intrinsic AKI. However, the combination of dehydration with advanced age or hypertension induces AKF evidenced by a magnified response of renal dysfunction (reduced filtration and tubular function) and tubular necrosis caused by low-dose cisplatin treatment. This study highlights the relevance of addressing AKF as a premorbid condition providing prophylactic opportunities and shows that dehydration differentially predisposes to prerenal and intrinsic AKI.</p>","PeriodicalId":17930,"journal":{"name":"Laboratory Investigation","volume":" ","pages":"102211"},"PeriodicalIF":5.1,"publicationDate":"2024-12-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142829284","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-13DOI: 10.1016/j.labinv.2024.102209
Miriam Mengoni, Felix O Mahlo, Evelyn Gaffal, Thomas Tüting, Andreas D Braun
The success of immune checkpoint inhibitors (ICI) in melanoma therapy has catalyzed the introduction of ICI in increasingly early stages of the disease. This exposes many patients with a lower risk of relapse to the risk of protracted adverse events, highlighting the need for biomarkers guiding the use of ICI. Already many years ago, brisk infiltration of primary melanomas by lymphocytes has been linked to improved patient outcome, but controversial findings due to a high variability in classification systems have been described CD8+ T cells have been identified as a primary mediator of antitumor immunity in patients treated with ICI. As CD8+ T cells require the presentation of antigens via MHC-I on target cells, downregulation and loss of MHC-I have been observed as resistance mechanisms to ICI. In this study, we revisit the role of MHC-I expression and CD8+ T-cell infiltration in melanoma evolution using a cohort of advanced primary and matched metastatic melanomas by using an automated immunohistochemistry and digital pathology workflow. Our results show that downregulation of MHC-I expression is a frequent event in advanced primary melanomas that is associated with decreased CD8+ T-cell infiltration and an early metastatic spread to sentinel lymph nodes. Furthermore, MHC-I downregulation and decreased infiltration with CD8+ T cells are also associated with resistance to ICI. Our results suggest that analyses of MHC-I expression and CD8+ T-cell infiltration patterns could serve as future biomarkers to guide the decision to treat patients in early stages of melanoma with ICI.
免疫检查点抑制剂(ICI)在黑色素瘤治疗中取得的成功促使越来越多的早期患者开始接受 ICI 治疗。这使得许多复发风险较低的患者面临长期不良反应的风险,从而突出了对指导使用 ICI 的生物标志物的需求。早在多年前,原发性黑色素瘤的淋巴细胞快速浸润就被认为与患者预后的改善有关,但由于分类系统差异较大,研究结果存在争议。在接受 ICI 治疗的患者中,CD8+ T 细胞已被确定为抗肿瘤免疫的主要介质。由于 CD8+ T 细胞需要通过靶细胞上的 MHC-I 呈递抗原,因此已观察到 MHC-I 的下调和缺失是 ICI 的抵抗机制。在这里,我们使用自动免疫组化和数字病理学工作流程,通过一组晚期原发性和匹配的转移性黑色素瘤,重新审视了 MHC-I 表达和 CD8+ T 细胞浸润在黑色素瘤演变中的作用。我们的研究结果表明,MHC-I表达下调是晚期原发性黑色素瘤的常见现象,它与CD8+ T细胞浸润减少和向前哨淋巴结的早期转移扩散有关。此外,MHC-I 下调和 CD8+ T 细胞浸润减少还与对 ICI 的耐药性有关。我们的研究结果表明,对MHC-I表达和CD8+ T细胞浸润模式的分析可作为未来的生物标志物,指导对黑色素瘤早期患者进行ICI治疗的决策。
{"title":"Downregulation of MHC-I on Melanoma Cells and Decreased CD8+ T-Cell Infiltration Are Associated With Metastatic Spread and Resistance to Immunotherapy.","authors":"Miriam Mengoni, Felix O Mahlo, Evelyn Gaffal, Thomas Tüting, Andreas D Braun","doi":"10.1016/j.labinv.2024.102209","DOIUrl":"10.1016/j.labinv.2024.102209","url":null,"abstract":"<p><p>The success of immune checkpoint inhibitors (ICI) in melanoma therapy has catalyzed the introduction of ICI in increasingly early stages of the disease. This exposes many patients with a lower risk of relapse to the risk of protracted adverse events, highlighting the need for biomarkers guiding the use of ICI. Already many years ago, brisk infiltration of primary melanomas by lymphocytes has been linked to improved patient outcome, but controversial findings due to a high variability in classification systems have been described CD8+ T cells have been identified as a primary mediator of antitumor immunity in patients treated with ICI. As CD8+ T cells require the presentation of antigens via MHC-I on target cells, downregulation and loss of MHC-I have been observed as resistance mechanisms to ICI. In this study, we revisit the role of MHC-I expression and CD8+ T-cell infiltration in melanoma evolution using a cohort of advanced primary and matched metastatic melanomas by using an automated immunohistochemistry and digital pathology workflow. Our results show that downregulation of MHC-I expression is a frequent event in advanced primary melanomas that is associated with decreased CD8+ T-cell infiltration and an early metastatic spread to sentinel lymph nodes. Furthermore, MHC-I downregulation and decreased infiltration with CD8+ T cells are also associated with resistance to ICI. Our results suggest that analyses of MHC-I expression and CD8+ T-cell infiltration patterns could serve as future biomarkers to guide the decision to treat patients in early stages of melanoma with ICI.</p>","PeriodicalId":17930,"journal":{"name":"Laboratory Investigation","volume":" ","pages":"102209"},"PeriodicalIF":5.1,"publicationDate":"2024-12-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142829286","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}