Michelle A Anderson’s research while affiliated with University of Michigan and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (198)


Metal Biliary Stents in Benign Pancreaticobiliary Disease
  • Chapter

May 2024

Michelle A. Anderson

·


Epithelial subtyping reveals subpopulations of basal, classical, and intermediary cancer cells. A, Uniform Manifold Approximation and Projection (UMAP) of scRNA-seq of 18 treatment-naïve PDAC. B, UMAP of tumor epithelial cells extracted from scRNA-seq of PDAC. Numbers represent unbiased clustering of populations. C, Dotplot showing average expression and percent cells of top expressing markers in each epithelial cluster. Cluster 5 markers outlined in green box. Genes highlighted yellow represent classical subtype markers. Genes outlined in magenta represent basal subtype markers. D, Feature plots showing expression of CLDN18, GATA6, KRT17, and CXCL8 in tumor epithelial cells. Cluster 5 epithelial cells denoted by dashed oval. E, Feature plots of gene set scoring of classical and basal signatures on tumor epithelial cells. F, Gene set scoring of each epithelial cluster for classical and basal gene sets. Numbers represent unbiased clustering of epithelial populations. Blue arrow denotes epithelial Cluster 5 (cluster with highest expression of KRT17, CLDN18, and CXCL8). G, Violin plots showing normalized expression of top expressing markers from epithelial Cluster 5 within the basal clusters (Clusters 3, 8, and 10), classical clusters (Clusters 0 and 7), and intermediary clusters (Clusters 5 and 6). H, Functional annotation showing top pathways expressed in epithelial Cluster 5 utilizing KEGG. The size of each dot represents gene count.
CXCL8⁺ tumor prevalence is associated with worse OS. A, Surgically resected PDAC tissue stained with combined antibody immunostaining for KRT17 (white) and ISH utilizing a probe for CXCL8 (green) and CXCL1 (red). B, Surgically resected PDAC tissue stained with combined antibody immunostaining for KRT17 (green)/CLDN18 (red) and ISH utilizing a probe for CXCL8 (magenta). White arrow denotes cell that is positive for CXCL8, KRT17, and CLDN18. C, Histogram of single-cell epithelial cluster frequencies by patient. D, Multivariate survival analysis of Puleo cohort using significant covariates from univariate survival analysis of top expressing markers from epithelial Cluster 5 and clinical features as variables. Pink highlighted genes represent significant increase in hazard while blue highlighted genes represent significant decrease in hazard. E, Kaplan–Meier curve of Puleo cohort of patients with high versus low IL8 tumoral gene expression (determined by top and bottom quartile), n = 288.
CXCL8 is expressed in a unique subpopulation of tumor epithelial cells and in tumor-infiltrating granulocytes. A, Surgically resected PDAC stained with combined antibody immunostaining for epithelial cells (E-cadherin, white), immune cells (CD45, red), and ISH utilizing a probe for CXCL8 (green). B, Three surgically resected treatment-naïve PDAC (left) and three surgically resected treated PDAC (right) stained with combined antibody immunostaining for epithelial cells (E-cadherin, white), immune cells (CD45, red), and ISH utilizing a probe for CXCL8 (green).
Functional analysis ex vivo reveals reciprocal interactions between cancer cells and granulocytes in the pancreatic cancer microenvironment mediated by CXCL8. A, Light microscopy (left) and H&E staining (right) of organoid lines isolated from tissue of 5 patients with PDAC. B, Organoids stained with antibody immunostaining for KRT17 (white) and ISH utilizing a probe for CXCL8 (green) and CXCL1 (red). C, Scheme of coculture to assay chemotaxis of PDAC patient myeloid cells to PDAC organoid conditioned media. D, Migration assay showing chemotaxis index (ratio of cells migrated through transwell in treatment condition compared with cells migrated in control media) in organoid lines with and without CXCL8-blocking antibody. Recombinant CXCL8 (rhCXCL8) was used as a positive control. Comparisons were made using one-way ANOVA. E, Scheme of assay to profile CXCL8 levels in healthy donor CD11b⁺ myeloid cells in response to PDAC organoid conditioned media. F, Relative fold change in CXCL8 mRNA levels in myeloid cells isolated from healthy donor blood treated with conditioned media in CXCL8high versus CXCL8low tumor organoid lines (1225 and 1253) with and without anti CXCL8-blocking antibody.
Tumor-derived CXCL8 correlates with specific changes in the local and systemic immune system. A, Scheme of workflow to compare scRNA-seq of matched tumor-infiltrating granulocytes from patients with tumors high in CXCL8⁺ epithelial cells versus tumors low in CXCL8⁺ epithelial cells. B, Unbiased differential expression between tumor-infiltrating granulocytes from tumors high in CXCL8⁺ epithelial cells versus tumors low in CXCL8⁺ epithelial cells. Significantly upregulated and downregulated genes are plotted as the average expression. Purple arrows denote genes associated with protumor phenotype, upregulated in epithelial CXCL8high group. Blue arrows denote upregulation of CXCL8 receptors CXCR1 and CXCR2 in epithelial CXCL8low group. C, UMAP visualization of myeloid cells extracted from PDAC scRNA-seq. D, Average expression of CXCL8 in the epithelial and myeloid subsets by patient. E, Scatterplot of percent positive CXCL8 myeloid cells versus percent positive CXCL8 epithelial cells. Each dot represents one patient. F, Feature plot of CXCL8 in tumor-infiltrating myeloid cells (left). Extracted granulocyte cells, colored by cluster (right). G, Top expressed genes within granulocyte Clusters G1 through G5. H, Violin plots showing normalized expression of select markers in granulocyte Clusters G1 through G5. I, Scheme of workflow to compare scRNA-seq of matched peripheral blood granulocytes from patients with tumors high in CXCL8⁺ epithelial cells versus tumors low in CXCL8⁺ epithelial cells. J, Unbiased differential expression between peripheral blood granulocytes from patients with tumors high in CXCL8⁺ epithelial cells versus tumors low in CXCL8⁺ epithelial cells. Significantly upregulated and downregulated genes are plotted as the average expression. Purple arrows denote genes associated with protumor phenotype, upregulated in epithelial CXCL8high group. Blue arrows denote upregulation of genes associated with antitumor phenotype, upregulated in epithelial CXCL8low group. K, UMAP visualization of all captured cells from PDAC blood of our single-cell cohort. Populations defined by color. L, Top: UMAP visualization of extracted granulocytes from PDAC blood single-cell sequencing. Numbers represent unbiased clustering of populations. Bottom: CXCL8 feature plot of extracted granulocytes. M, Dotplot showing average expression and percent cells expressed of CXCL8, CXCR1, CXCR2, CXCR4, SPP1, CCL4, SELL, and VEGFA in each peripheral blood granulocyte cluster. Green box outlines Cluster 0, the cluster with highest expression of CXCL8.

+3

KRT17/CXCL8 Tumor Cells Display Both Classical and Basal Features and Regulate Myeloid Infiltration in the Pancreatic Cancer Microenvironment
  • Article
  • Full-text available

October 2023

·

120 Reads

·

16 Citations

Eileen S. Carpenter

·

Padma Kadiyala

·

Ahmed M. Elhossiny

·

[...]

·

Purpose Pancreatic ductal adenocarcinoma (PDAC) is generally divided in two subtypes, classical and basal. Recently, single-cell RNA sequencing has uncovered the coexistence of basal and classical cancer cells, as well as intermediary cancer cells, in individual tumors. The latter remains poorly understood; here, we sought to characterize them using a multimodal approach. Experimental Design We performed subtyping on a single-cell RNA sequencing dataset containing 18 human PDAC samples to identify multiple intermediary subtypes. We generated patient-derived PDAC organoids for functional studies. We compared single-cell profiling of matched blood and tumor samples to measure changes in the local and systemic immune microenvironment. We then leveraged longitudinally patient-matched blood to follow individual patients over the course of chemotherapy. Results We identified a cluster of KRT17-high intermediary cancer cells that uniquely express high levels of CXCL8 and other cytokines. The proportion of KRT17high/CXCL8⁺ cells in patient tumors correlated with intratumoral myeloid abundance, and, interestingly, high protumor peripheral blood granulocytes, implicating local and systemic roles. Patient-derived organoids maintained KRT17high/CXCL8⁺ cells and induced myeloid cell migration in a CXCL8-dependent manner. In our longitudinal studies, plasma CXCL8 decreased following chemotherapy in responsive patients, while CXCL8 persistence portended worse prognosis. Conclusions Through single-cell analysis of PDAC samples, we identified KRT17high/CXCL8⁺ cancer cells as an intermediary subtype, marked by a unique cytokine profile and capable of influencing myeloid cells in the tumor microenvironment and systemically. The abundance of this cell population should be considered for patient stratification in precision immunotherapy. See related commentary by Faraoni and McAllister, p. 2297

Download

Acute pancreatitis precedes chronic pancreatitis in the majority of patients: Results from the NAPS2 consortium

October 2022

·

107 Reads

·

18 Citations

Pancreatology

Introduction The mechanistic definition of chronic pancreatitis (CP) identifies acute pancreatitis (AP) as a precursor stage. We hypothesized that clinical AP frequently precedes the diagnosis of CP and is associated with patient- and disease-related factors. We describe the prevalence, temporal relationship and associations of AP in a well-defined North American cohort. Methods We evaluated data from 883 patients with CP prospectively enrolled in the North American Pancreatitis Studies across 27 US centers between 2000 and 2014. We determined how often patients had one or more episodes of AP and its occurrence in relationship to the diagnosis of CP. We used multivariable logistic regression to determine associations for prior AP. Results There were 624/883 (70.7%) patients with prior AP, among whom 161 (25.8%) had AP within 2 years, 115 (18.4%) within 3–5 years, and 348 (55.8%) >5 years prior to CP diagnosis. Among 504 AP patients with available information, 436 (86.5%) had >1 episode. On multivariable analyses, factors associated with increased odds of having prior AP were a younger age at CP diagnosis, white race, abdominal pain, pseudocyst(s) and pancreatic duct dilatation/stricture, while factors associated with a lower odds of having prior AP were exocrine insufficiency and pancreatic atrophy. When compared with patients with 1 episode, those with >1 AP episode were diagnosed with CP an average of 5 years earlier. Conclusions Nearly three-quarters of patients were diagnosed with AP prior to CP diagnosis. Identifying which AP patients are at-risk for future progression to CP may provide opportunities for primary and secondary prevention.




Abstract PO-098: Longitudinal profiling of pancreatic cancer patients identifies interleukin-8 as a mediator of myeloid-epithelial crosstalk

November 2021

·

51 Reads

Background: Pancreatic ductal adenocarcinoma (PDAC) is the 4th leading cause of cancer-related death in the US. A key hallmark of this disease is that, while tumors initially show susceptibility to standard chemotherapeutic agents, most patients eventually develop resistance, leading to poor survival. While the mechanisms of chemoresistance are unclear, murine studies have implicated the myeloid compartment of the tumor immune microenvironment. Correlative data in human tumors supports this notion, however, mechanistic studies are lacking, thus impairing translation to the clinic. The study of human pancreatic cancer has historically been challenging due to difficulty of fresh biospecimen acquisition, patient heterogeneity, and a diverse tumor microenvironment. Moreover, the vast majority of pancreatic cancer patients do not qualify for surgical resection, further limiting tissue availability. We have overcome these difficulties by developing a pipeline to analyze human tumor samples and matched blood using high-fidelity techniques including single-cell RNA sequencing (scRNAseq) and mass cytometry (CyTOF), together with establishment of organoids from the same tumors. Notably, in this pipeline we can use small amounts of tissue from endoscopic fine needle biopsies, thus allowing us to sample tumors from patients at any disease stage. Results: We performed CyTOF on longitudinally-matched peripheral blood mononuclear cells (PBMCs) from 30 patients and single-cell RNA sequencing on 6 patients in the treatment naïve and on-treatment (FOLFIRINOX) state. CyTOF revealed distinct alterations in the myeloid population, with a shift toward CXCR2hiPD-L1hi granulocytes with FOLFIRINOX treatment over time. Analysis of PBMCs from scRNAseq showed a distinct myeloid gene signature with FOLFIRINOX and in particular highlighted interleukin-8 (IL8), a chemokine involved in myeloid cell chemotaxis that is associated with poor prognosis in pancreatic cancer. Further mapping of IL8 in tumor tissue by scRNAseq showed that it is highly expressed in subpopulations of tumor epithelial cells and tumor-infiltrating granulocytes. IL8-high tumor-infiltrating granulocytes also highly expressed VEGF and CXCR4, suggesting immunosuppressive and angiogenic roles. IL8-high tumor epithelial cells were found to have a basal-like phenotype and also expressed a network of other chemokines including CXCL1, CXCL3, CXCL5, which are known to recruit immunosuppressive myeloid cells. Conclusions: Through longitudinal and multimodal mapping using PDAC patient blood and tumor biospecimens, we have identified IL8 as a potential mediator of epithelial-myeloid crosstalk in PDAC chemoresistance and tumor aggression. Validation studies using an all-human co-culture system of PDAC patient-derived organoids and myeloid cells are currently underway. Citation Format: Eileen S. Carpenter, Samantha Kemp, Padma Kadiyala, Nina Steele, Ahmed Elhossiny, Stephanie The, Valerie Gunchick, Rémy Nicolle, Michelle Anderson, Wenting Du, Carlos Espinoza, Richard Kwon, Erik-Jan Wamsteker, Anoop Prabhu, Allison Schulman, Vaibhav Sahai, Timothy Frankel, Filip Bednar, Marina Pasca di Magliano. Longitudinal profiling of pancreatic cancer patients identifies interleukin-8 as a mediator of myeloid-epithelial crosstalk [abstract]. In: Proceedings of the AACR Virtual Special Conference on Pancreatic Cancer; 2021 Sep 29-30. Philadelphia (PA): AACR; Cancer Res 2021;81(22 Suppl):Abstract nr PO-098.



Chief of Endoscopy: Specific Challenges to Leading the Team and Running the Unit

April 2021

·

10 Reads

Techniques and Innovations in Gastrointestinal Endoscopy

Healthcare organizations have recognized the value of physician leaders in change management and achieving institutional goals. Physicians leaders are now at all levels from individual clinics to the very top of large, multi-hospital organizations. Most endoscopy units have medical directors and organizations with multiple or large, complex units often have a “Chief of Endoscopy” (CoE) or the equivalent of this to oversee and lead the teams and decisions pertaining to these units. The following review will discuss the process to become a CoE, the importance of aligning personal values and skills with organizational purpose and will give suggestions for relevant metrics to success. It will also explore the challenges faced by women in endoscopy leadership positions and will give advice to women leaders and those who support her as mentors, colleagues, or partners. Although much of the discussion is intentionally focused on high-level endoscopy leadership, the information and messages have pertinence to any organizational leadership position and to leaders at any point in their leadership pathway.


Potential facilitators to women pursuing a career in advanced endoscopy.
Fraction of female graduates over the past 10 years is positively associated with fraction of female advanced endoscopy faculty (ß = 0.43, P < 0.001).
Gender disparities in advanced endoscopy fellowship

February 2021

·

132 Reads

·

29 Citations

Background and study aims Women remain underrepresented in gastroenterology, especially advanced endoscopy. Women represent 30 % of general gastroenterology fellows; yet in 2019, only 12.8 % of fellows who matched into advanced endoscopy fellowship (AEF) programs were women. Methods We administered a web-based survey to the program directors (PDs) of AEF programs that participated in the 2018–2019 American Society for Gastroenterology (ASGE) match. We assessed PD and program characteristics, in addition to perceived barriers and facilitators (scale 1–5, 5 = most important) influencing women pursuing AEF training. Results We received 38 (59.3 %) responses from 64 PDs. 15.8 % (6/38) of AEF PDs and 13.2 % (5/38) of endoscopy chiefs were women. By program, women represented 14.8 % (mean) ± 17.0 % (SD) of AEF faculty and 12.0 % (mean) ± 11.1 % (SD) of AEF trainees over the past 10 years. 47.4 % (18/38) programs reported no female advanced endoscopy faculty and 31.6 % (12/38) of programs have never had a female fellow. Percentage of female fellows was strongly associated with percentage of female AEF faculty (ß = 0.43, P < 0.001). Inflexible hours and call (mean rank 3.3 ± 1.1), exposure to fluoroscopy (2.9 ± 1.1), lack of women endoscopists at national conferences/courses (2.9 ± 1.1) and lack of female mentorship (2.9 ± 1.0) were cited as the most important barriers to recruitment. Conclusion We utilized a survey of AEF PDs participating in the ASGE match to determine program characteristics and identify contributors to gender disparity. Women represent a minority of AEF PDs, endoscopy chiefs, advanced endoscopy faculty and AEF trainees. Our study highlights perceived barriers and facilitators to recruitment, and emphasizes the importance of having female representation in faculty, and leadership positions in endoscopy.


Hedgehog pathway activation is higher in myCAFs compared with iCAFs in PDAC. A, UMAP visualization of cell populations from scRNA-seq of human adjacent normal/normal pancreas (n = 3) and PDAC (n = 16) tissues. The normal samples (n = 3) were isolated from patients undergoing surgery for duodenal adenoma, ampullary carcinoma, or PDAC, where an uninvolved portion of the pancreas was included in the resection. Different cell type clusters are color coded. Data are from Steele and colleagues (35). B, Dot plot visualization of Hedgehog pathway gene expression level (color intensity) and frequency (size of dot) in different cell populations of human adjacent normal/normal pancreas (blue, n = 3) and PDAC (red, n = 16) samples from A. Boxes highlight Hedgehog ligands (SHH, IHH, and DHH) and Hedgehog targets (GLI1, PTCH1, PTCH2, and HHIP). C, UMAP visualization of scRNA-seq of the fibroblast clusters in pooled human adjacent normal pancreas (n = 2) and PDAC (n = 6) samples. Different CAF subtype clusters are color coded. Data are from Elyada and colleagues (30). D, UMAP visualization of myCAF (ACTA2 coding for αSMA and TAGLN) and iCAF (IL6 and CLEC3B) marker expression in the fibroblast clusters in human PDAC samples from C. E, UMAP visualization of Hedgehog target (GLI1 and PTCH1) expression in the fibroblast clusters in human PDAC samples from C. F, Heatmaps of normalized expression of Hedgehog targets (GLI1, PTCH1, PTCH2, and HHIP), Hedgehog receptor (SMO), and coreceptors (GAS1, CDON, and BOC) in each fibroblast cluster in human PDAC samples from C. Colors indicate log-scale gene counts. G, Heatmap of scaled expression of Hedgehog ligands (Shh, Ihh, and Dhh), Hedgehog targets (Gli1, Ptch1, Ptch2, and Hhip), Hedgehog receptor (Smo), and coreceptors (Gas1, Cdon, and Boc) in different cell populations of pancreatic tumors of the KPC mouse model of PDAC (n = 4). Data are scaled such that the cluster with the lowest average expression = 0 and the highest = 1 for each gene. Data are from Elyada and colleagues (30). H, Representative RNA ISH of Gli1 (white) and co-IF of PDPN (green) in a KPC tumor. Counterstain, DAPI (blue). Scale bar, 20 μm. I, Quantitation of Gli1 stain in PDPN⁺ (CAFs) and PDPN⁻ (non-CAFs) cells in KPC tumors. Results show mean ± SEM of seven biological replicates. ***, P < 0.001, unpaired Student t test. J, UMAP visualization of scRNA-seq of the fibroblast clusters in KPC tumors (n = 4) from Elyada and colleagues (30). Different CAF subtype clusters are color coded. K, Heatmaps of normalized expression of Hedgehog targets (Gli1, Ptch1, Ptch2, and Hhip), Hedgehog receptor (Smo), and coreceptors (Gas1, Cdon, and Boc) in each fibroblast cluster from J. Colors indicate log-scale gene counts. L, Representative RNA ISH of Gli1 (white) and co-IF of PDPN (green) and αSMA (red) in a KPC tumor. Counterstain, DAPI (blue). Scale bar, 20 μm. The arrowhead points at a PDPN⁺αSMA⁺ cell with lower Gli1 expression; solid arrow points at a PDPN⁺αSMA⁺ cell with higher Gli1 expression. M, Quantitation of Gli1 stain in αSMA⁺ PDPN⁺ (myCAFs) and αSMA⁻ PDPN⁺ (non-myCAFs) cells in KPC tumors. Results show mean ± SEM of seven biological replicates. **, P < 0.01, unpaired Student t test.
Hedgehog pathway inhibition impairs PDAC growth. A, Schematic of orthotopic transplantation of 7940b KPC PDAC cells with Ihh WT or KO in BL/6J or Gli1lacZ/+ mice. B, Tumor weights at day 18 after transplantation of the experiment from A. Results show mean ± SEM of six biological replicates. **, P < 0.01, one-way ANOVA. C, Representative RNA ISH images of Gli1 in Ihh WT or -KO tumors in BL/6J mice (left), and of X-GAL stain and co-IF of αSMA (magenta), β-gal (green), and DAPI (blue) in Ihh WT or -KO tumors in Gli1lacZ/+ mice (middle and right). Inserts, magnifications. Scale bars, 50 μm (left), 100 μm (middle), and 25 μm (right). D, Quantitation of Gli1 RNA ISH in Ihh WT or -KO tumors in BL/6J mice. Results show mean ± SEM on six biological replicates. ***, P < 0.001, unpaired Student t test. E, Quantitation of X-GAL stain in Ihh WT or -KO tumors in Gli1lacZ/+ mice. Results show mean ± SEM on six biological replicates. **, P < 0.01, unpaired Student t test. F, Schematic of orthotopic transplantation of 7940b KPC PDAC cells into BL/6J or Gli1lacZ/+ mice, followed by a 12-day treatment with 20 mg/kg SMO inhibitor, LDE225, or vehicle by daily oral gavage. G, Tumor weights at day 18 after transplantation of the experiment from F. Results show mean ± SEM of 4–6 biological replicates. ***, P < 0.001, one-way ANOVA. H, Representative RNA ISH images of Gli1 in vehicle- or LDE225-treated tumors in BL/6J mice (left), and of X-GAL stain and co-IF of αSMA (red), β-gal (green), and DAPI (blue) in vehicle- or LDE225-treated tumors in Gli1lacZ/+ mice (middle and right). Inserts, magnifications. Scale bars, 50 μm (left), 100 μm (middle), and 25 μm (right). I, Quantitation of Gli1 RNA ISH in LDE225- or vehicle-treated tumors in BL/6J mice. Results show mean ± SEM on four biological replicates. ***, P < 0.001, unpaired Student t test. J, Quantitation of X-GAL stain in LDE225- or vehicle- treated tumors in Gli1lacZ/+ mice. Results show mean ± SEM on four biological replicates. ***, P < 0.001, unpaired Student t test. K, Schematic of 2-week treatment of tumor-bearing KPC mice with 20 mg/kg LDE225 or vehicle by daily oral gavage. U/S, ultrasound. L, Tumor volume as measured by ultrasound of vehicle- (n = 9) and LDE225-treated (n = 8) KPC tumors from K. Results show mean ± SEM. *, P < 0.05, unpaired Student t test. M, Representative RNA ISH of Gli1 in 2-week vehicle- (n = 11) and LDE225-treated (n = 10) KPC tumors. Scale bar, 100 μm. N, Quantitation of Gli1 stain in vehicle- (n = 11) and LDE225-treated (n = 10) KPC tumors. Results show mean ± SEM. ***, P < 0.001, unpaired Student t test.
Two-week Hedgehog pathway inhibition alters the fibroblast compartment in PDAC. A, Representative IHC of PDPN in vehicle- (n = 10) and LDE225-treated (n = 11) KPC tumors. Scale bar, 50 μm. B, Quantitation of PDPN stain in vehicle- (n = 10) and LDE225-treated (n = 11) KPC tumors. Results show mean ± SEM. **, P < 0.01, unpaired Student t test. C, Schematic of FACS strategy for bulk RNA-seq of fibroblasts from vehicle- and LDE225-treated KPC tumors. D, GSEA of cell-cycle signature in FACS-sorted CAFs from LDE225-treated KPC tumors (n = 2) compared with FACS-sorted CAFs from vehicle-treated controls (n = 3). E, RNA-seq expression of proliferation markers (Mki67, Ccnb2, and Cks2) in FACS-sorted CAFs from vehicle- (n = 3) and LDE225-treated (n = 2) KPC tumors. Results show mean ± SEM. *, P < 0.05; **, P < 0.01; ***, P < 0.001, unpaired Student t test. F, Co-IF of αSMA (red) and PDPN (green) in vehicle- (n = 7) and LDE225-treated (n = 4) KPC tumors. Counterstain, DAPI. Scale bar, 20 μm. G, Quantitation of myCAFs (αSMA⁺ PDPN⁺ DAPI⁺) in vehicle- (n = 7) and LDE225-treated (n = 4) KPC tumors. Results show mean ± SEM. P = 0.05, unpaired Student t test. H, Quantitation of non-myCAFs (αSMA⁻ PDPN⁺ DAPI⁺) in vehicle- (n = 7) and LDE225-treated (n = 4) KPC tumors. Results show mean ± SEM. *, P < 0.05, unpaired Student t test.
Two-week Hedgehog pathway inhibition alters the ratio of myCAFs and iCAFs in PDAC. A, Schematic of flow cytometry strategy for myCAFs and iCAFs from 2-week vehicle- and LDE225-treated KPC tumors. B, Representative flow plots showing the gating strategy for the analysis of DAPI⁻ CD45⁻ CD31⁻ EpCAM⁻ PDPN⁺ CAFs in 2-week vehicle- (n = 7) and LDE225-treated (n = 6) KPC tumors. C, Flow cytometry analysis of myCAFs (calculated from DAPI⁺ singlets) in vehicle- (n = 7) and LDE225-treated (n = 6) KPC tumors. Results show mean ± SEM. Unpaired Student t test. D, Flow cytometry analysis of iCAFs (calculated from DAPI⁺ singlets) in vehicle- (n = 7) and LDE225-treated (n = 6) KPC tumors. Results show mean ± SEM. *, P < 0.05, unpaired Student t test. E, Proportions of myCAF, iCAF, and apCAF subtypes from the PDPN⁺ gate in vehicle- (n = 7, top) and LDE225-treated (n = 6, bottom) KPC tumors, as measured by flow cytometry analysis. Results show average percentage of biological replicates. F, Flow cytometric analysis of the iCAF/myCAF ratio from the PDPN⁺ gate in vehicle- (n = 7) and LDE225-treated (n = 6) KPC tumors. Results show mean ± SEM. *, P < 0.05, unpaired Student t test. G, GSEA of the myCAF gene signature in FACS-sorted CAFs from 2-week LDE225-treated KPC tumors (n = 2) compared with FACS-sorted CAFs from vehicle-treated controls (n = 3). The myCAF gene signature was defined from the study by Öhlund and colleagues (27). H, RNA-seq expression of myCAF markers (Acta2, Thy1, Tagln, and Tgfb1) in FACS-sorted CAFs from vehicle- (n = 3) and LDE225-treated (n = 2) KPC tumors. Results show mean ± SEM. No statistical difference was observed as calculated by unpaired Student t test. I, GSEA of the iCAF gene signature in FACS-sorted CAFs from 2-week LDE225-treated KPC tumors (n = 2) compared with FACS-sorted CAFs from vehicle-treated controls (n = 3). The iCAF gene signature was defined from the study by Öhlund and colleagues (27). J, RNA-seq expression of iCAF markers (Dpt, Clec3b, C3, and Cxcl12) in FACS-sorted CAFs from vehicle- (n = 3) and LDE225-treated (n = 2) KPC tumors. Results show mean ± SEM. *, P < 0.05, unpaired Student t test.
Two-week Hedgehog (HH) pathway inhibition alters the immune infiltration in pancreatic tumors. A, Schematic of 2-week treatment of tumor-bearing KPC mice with 20 mg/kg LDE225 or vehicle prior to CyTOF analysis (top). U/S, ultrasound. Table of CyTOF panel including metal tag, antibody target, and cell type predominantly expressed (bottom). See Supplementary Table S1 for detailed antibody information. B, Manual gating of CyTOF data for total myeloid cells (CD45⁺CD11b⁺), macrophages (CD11b⁺F4/80⁺), PDL1⁺ macrophages (F4/80⁺PD-L1⁺), and CD206⁺ macrophages (F4/80⁺CD206⁺) in 2-week vehicle- (n = 7) and LDE225-treated (n = 6) KPC tumors. Results show mean ± SEM. *, P < 0.05, unpaired Student t test. C, Manual gating of CyTOF data for CD8⁺, CD4⁺, and CD4⁺CD25⁺ T cells as a percentage of total CD3⁺ T cells. Results show mean ± SEM. *, P < 0.05; **, P < 0.01, unpaired Student t test. D, Representative IHC of CD8A in 2-week vehicle- and LDE225-treated KPC tumors. Inserts, magnifications. Scale bar, 50 μm. E, Quantitation of CD8A stain in 2-week vehicle- (n = 12) and LDE225-treated (n = 11) KPC tumors. Results show mean ± SEM. *, P < 0.05, unpaired Student t test. F, Representative IHC of FOXP3 in 2-week vehicle- and LDE225-treated KPC tumors. Inserts, magnifications. Scale bar, 50 μm. G, Quantitation of FOXP3 stain in 2-week vehicle- (n = 12) and LDE225-treated (n = 11) KPC tumors. Results show mean ± SEM. **, P < 0.01, unpaired Student t test. H, Model explaining the role of Hedgehog signaling and the effects of Hedgehog inhibition on the PDAC microenvironment. Cancer-secreted Hedgehog ligands, such as SHH and IHH, activate Hedgehog signaling in surrounding fibroblasts (arrow), especially in myCAFs (left). Hedgehog inhibition leads to a reduction in myCAFs and an increase in iCAFs, and to decreased CD8⁺ T cells and more abundant regulatory T cells (right).
Inhibition of Hedgehog Signaling Alters Fibroblast Composition in Pancreatic Cancer

January 2021

·

296 Reads

·

227 Citations

Purpose Pancreatic ductal adenocarcinoma (PDAC) is a deadly disease characterized by an extensive fibroinflammatory stroma, which includes abundant cancer-associated fibroblast (CAF) populations. PDAC CAFs are heterogeneous, but the nature of this heterogeneity is incompletely understood. The Hedgehog pathway functions in PDAC in a paracrine manner, with ligands secreted by cancer cells signaling to stromal cells in the microenvironment. Previous reports investigating the role of Hedgehog signaling in PDAC have been contradictory, with Hedgehog signaling alternately proposed to promote or restrict tumor growth. In light of the newly discovered CAF heterogeneity, we investigated how Hedgehog pathway inhibition reprograms the PDAC microenvironment. Experimental Design We used a combination of pharmacologic inhibition, gain- and loss-of-function genetic experiments, cytometry by time-of-flight, and single-cell RNA sequencing to study the roles of Hedgehog signaling in PDAC. Results We found that Hedgehog signaling is uniquely activated in fibroblasts and differentially elevated in myofibroblastic CAFs (myCAF) compared with inflammatory CAFs (iCAF). Sonic Hedgehog overexpression promotes tumor growth, while Hedgehog pathway inhibition with the smoothened antagonist, LDE225, impairs tumor growth. Furthermore, Hedgehog pathway inhibition reduces myCAF numbers and increases iCAF numbers, which correlates with a decrease in cytotoxic T cells and an expansion in regulatory T cells, consistent with increased immunosuppression. Conclusions Hedgehog pathway inhibition alters fibroblast composition and immune infiltration in the pancreatic cancer microenvironment.


Citations (53)


... 35 Stress KRTs are known to be associated closely with the occurrence of various diseases, including psoriasis, cancer, and pachyonychia congenita. 8,[36][37][38][39][40] However, there is limited research on these important disease-related KRTs in ophthalmology. Some studies have shown that low expression of KRT16 plays an important role in highly myopic corneas and may be partly responsible for the lower corneal biomechanics in highly myopic eyes, 41 whereas in keratoconus patients, the expression of Krt16 is elevated. ...

Reference:

Expression Distribution of Keratins in Normal and Pathological Corneas and the Regulatory Role of Krt17 on Limbal Stem Cells
KRT17/CXCL8 Tumor Cells Display Both Classical and Basal Features and Regulate Myeloid Infiltration in the Pancreatic Cancer Microenvironment

... Genetic research into pancreatitis risk factors typically begins with studies on CP. However, it is now widely recognized that there is a continuum between acute pancreatitis (AP), recurrent AP (RAP), and CP [111][112][113]. To explain this progression, the sentinel acute pancreatitis event (SAPE) model was proposed [114]. ...

Acute pancreatitis precedes chronic pancreatitis in the majority of patients: Results from the NAPS2 consortium
  • Citing Article
  • October 2022

Pancreatology

... Consistently, reduced expression of the variant allele has been observed in two pancreatic cancer cell lines heterozygous for the SPINK1 p.Asn34Ser haplotype [61]. RNA transcript analyses from three individuals heterozygous for p.Asn34Ser similarly revealed significantly fewer transcript reads from the variant allele compared to the wildtype (WT) allele [62]. ...

Pancreatitis-associated PRSS1-PRSS2 haplotype alters T cell receptor beta (TRB) repertoire more strongly than PRSS1 expression
  • Citing Article
  • October 2022

Gastroenterology

... For example, in 2019 women represented only 14.0 % of all current advanced endoscopy fellows in the USA. [13] Barriers for women to achieve expertise in EUS and interventional endoscopy are also reported from Italy [14] and India. [15] These barriers include implicit gender bias, lack of flexibility in the training programs with regard to family planning and family obligations (e.g., part-time work options, maternity leave, hours and call), exposure to fluoroscopy during childbearing age, lack of ergonomic equipment, and lack of female program directors and educators. ...

Gender disparities in advanced endoscopy fellowship

... In PDAC, tumor cells secrete Hh ligand, while surrounding CAFs respond to these ligands and are subsequently activated [23]. Hh pathway activation impacts CAF states; inhibiting this pathway in a PDAC mouse model using the smoothened antagonist LDE225 was shown to reduce myCAF numbers while simultaneously enriching iCAFs, indicating a role of Hh signaling in shaping CAF phenotypes [24]. Other recent studies have shed light on the complex role of Hh pathway inhibition in PDAC and may explain the observed negative clinical outcomes. ...

Inhibition of Hedgehog Signaling Alters Fibroblast Composition in Pancreatic Cancer

... Inherent treatment resistance is a major challenge [11], and efforts to find better therapeutic strategies have only achieved incremental progress [8]. The importance of the tumor microenvironment (TME) for dictating responses to therapy and disease progression across different tumor entities is well established [1,[11][12][13][14]. Within the TME, cancer-associated fibroblasts (CAFs) deposit and remodel the extracellular matrix (ECM) [11,15], and immune cells establish an immunosuppressive milieu [12,16,17]. ...

Multimodal Mapping of the Tumor and Peripheral Blood Immune Landscape in Human Pancreatic Cancer

Nature Cancer

... Крім анамнестичних і клінічних даних, вивчено рівень сироваткового трипсиногену (ТпГ) та фекальної еластази-1 (ФЕ-1) за допомогою імуноферментних наборів «ELISA» виробництва «MyBio-Source» і «ScheBo ® Pancreatic Elastase 1™ Stool Test kit», відповідно до наданих до реактивів інструкцій. Пороговими значеннями концентрацій ферментів, нижче яких вже діагностували лабораторно підтверджену недостатність ПЗСА помірного ступеня, визнано показники <200 нг/мл для ФЕ-1 і <20 нг/мл для ТпГ [16]. ...

Low serum trypsinogen levels in chronic pancreatitis: Correlation with parenchymal loss, exocrine pancreatic insufficiency, and diabetes but not CT-based cambridge severity scores for fibrosis
  • Citing Article
  • September 2020

Pancreatology

... Hereditary pancreatitis, including cases associated with SPINK1 mutations, may lead to chronic inf lammation of the pancreas, increasing the risk of complications such as pancreatic insufficiency, diabetes, and an elevated risk of pancreatic cancer compared to the general population [6]. ...

Differences in Age at Onset of Symptoms, and Effects of Genetic Variants, in Patients With Early Vs Late-Onset Idiopathic Chronic Pancreatitis in a North American Cohort
  • Citing Article
  • March 2020

Clinical Gastroenterology and Hepatology

... TAMs in turn secrete immunosuppressive cytokines such as TGF-B and IL-10, among others, that hinder recruitment and activity of effector T cells, regulatory T cells, and cytotoxic CD8+ T-cells [93,94]. Various other immune cell subtypes in the PDAC TME, namely myeloid-derived suppressor cells, Th17 cells, and neutrophils, also contribute to its immunosuppressive nature [95][96][97][98][99][100][101][102][103][104]. Additionally, signaling protein VEGF plays a role in the TME, similarly impairing the recruitment and effector mechanisms of immune cells by interfering with angiogenesis [105]. ...

Regulatory T-cell Depletion Alters the Tumor Microenvironment and Accelerates Pancreatic Carcinogenesis

... As such, it is questionable whether this score could be applied in PDAC patients to identify patients with an even higher risk of VTE. Subgroup analyses regarding the performance of the Khorana score in PDAC patients have shown that the absolute 6-month incidence of VTE in patients with a high-risk Khorana score ranges from 12% (pulmonary embolism (PE) or deep venous thrombosis (DVT) only) to 41% (superficial vein thrombosis and abdominal VTE included); in one study, the Khorana score had an AUROC of 0.65 [19,[89][90][91][92]. A recent large populationbased cohort study showed that the Khorana score was unable to identify PDAC patients at a particularly high risk of VTE (HR = 1.03, 95% CI [0.66-1.61]) ...

Prediction of Venous Thromboembolism (VTE) in Patients with Pancreatic Cancer Using Clinical Data, Biomarkers, and VTE Risk Models
  • Citing Article
  • November 2012

Blood