Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters

Publication year range
1.
Nature ; 610(7931): 356-365, 2022 10.
Article in English | MEDLINE | ID: mdl-36198802

ABSTRACT

Hepatocellular carcinoma (HCC), the fourth leading cause of cancer mortality worldwide, develops almost exclusively in patients with chronic liver disease andĀ advanced fibrosis1,2. Here we interrogated functions of hepatic stellate cells (HSCs), the main source of liver fibroblasts3, during hepatocarcinogenesis. Genetic depletion, activation or inhibition of HSCs in mouse models of HCC revealed their overallĀ tumour-promoting role. HSCs were enriched in the preneoplastic environment, where they closely interacted with hepatocytes and modulated hepatocarcinogenesis by regulating hepatocyte proliferation and death. Analyses of mouse and human HSC subpopulations by single-cell RNA sequencing together with genetic ablation of subpopulation-enriched mediators revealed dual functions of HSCs in hepatocarcinogenesis. Hepatocyte growth factor, enriched in quiescent and cytokine-producing HSCs, protected against hepatocyte death and HCC development. By contrast, type I collagen, enriched in activated myofibroblastic HSCs, promoted proliferation and tumour development through increased stiffness and TAZ activation in pretumoural hepatocytes and through activation of discoidin domain receptor 1 in established tumours. An increased HSC imbalance between cytokine-producing HSCs and myofibroblastic HSCs during liver disease progression was associated with increased HCC risk in patients. In summary, the dynamic shift in HSC subpopulations and their mediators during chronic liver disease is associated with a switch from HCC protection to HCC promotion.


Subject(s)
Carcinogenesis , Carcinoma, Hepatocellular , Hepatic Stellate Cells , Liver Neoplasms , Animals , Carcinogenesis/pathology , Carcinoma, Hepatocellular/pathology , Cell Proliferation , Collagen Type I/metabolism , Discoidin Domain Receptor 1/metabolism , Disease Progression , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Hepatocyte Growth Factor/metabolism , Hepatocytes , Humans , Liver Cirrhosis/complications , Liver Neoplasms/pathology , Mice , Myofibroblasts/pathology
2.
Article in English | MEDLINE | ID: mdl-39278359

ABSTRACT

BACKGROUND: Severe hepatitis cases in children are increasingly recognized, but the exact etiology remains unknown in a significant proportion of patients. Cases of indeterminate severe hepatitis (iSH) may progress to indeterminate pediatric acute liver failure (iPALF), so understanding its immunobiology is critical to preventing disease progression. Hemophagocytic lymphohistiocytosis (HLH) is a systemic hyperinflammatory disorder associated with T-cell and macrophage activation with liver injury. OBJECTIVES: We hypothesized that a high proportion of patients with iSH demonstrate systemic T-cell activation similar to HLH before developing iPALF and that the degree of T-cell activation in iSH might correlate with outcomes. METHODS: From 2019 to 2022, 14 patients with iSH and 7 patients with PALF of known, nonimmune etiology were prospectively enrolled. We compared immune signatures of iSH, HLH, known PALF, and healthy controls. RESULTS: We found that patients with iSH have increased CD8+ T-cell activation and high IFN-ƎĀ³ activity similar to HLH. The amplitude of CD8+ T-cell activation was predictive of iSH progression to iPALF. We also found that in patients with iSH, ferritin had only modest elevation. However, the ratio of age-normalized plasma soluble IL-2 receptor to ferritin level can distinguish iSH from known PALF and HLH. As proof of concept, we report that in 3 patients with steroid-refractory iSH, emapalumab, an IFN-ƎĀ³ blocking antibody used in combination with steroids, improved liver function and may have prevented progression to PALF. CONCLUSIONS: Flow-based T-cell activation markers could help in early identification and risk stratification for targeted intervention in patients with iSH.

3.
J Hepatol ; 76(4): 910-920, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34902531

ABSTRACT

BACKGROUND & AIMS: Non-alcoholic steatohepatitis (NASH) is a leading cause of hepatocellular carcinoma (HCC), but mechanisms linking NASH to eventual tumor formation remain poorly understood. Herein, we investigate the role of TAZ/WWTR1, which is induced in hepatocytes in NASH, in the progression of NASH to HCC. METHODS: The roles of hepatocyte TAZ and its downstream targets were investigated in diet-induced and genetic models of NASH-HCC using gene-targeting, adeno-associated virus 8 (AAV8)-H1-mediated gene silencing, or AAV8-TBG-mediated gene expression. The biochemical signature of the newly elucidated pathway was probed in liver specimens from humans with NASH-HCC. RESULTS: When hepatocyte-TAZ was silenced in mice with pre-tumor NASH using AAV8-H1-shTaz (short-hairpin Taz), subsequent HCC tumor development was suppressed. In this setting, the tumor-suppressing effect of shTaz was not dependent of TAZ silencing in the tumors themselves and could be dissociated from the NASH-suppressing effects of shTaz. The mechanism linking pre-tumor hepatocyte-TAZ to eventual tumor formation involved TAZ-mediated induction of the NOX2-encoding gene Cybb, which led to NADPH-mediated oxidative DNA damage. As evidence, DNA damage and tumor formation could be suppressed by treatment of pre-tumor NASH mice with AAV8-H1-shCybb; AAV8-TBG-OGG1, encoding the oxidative DNA-repair enzyme 8-oxoguanine glycosylase; or AAV8-TBG-NHEJ1, encoding the dsDNA repair enzyme non-homologous end-joining factor 1. In surrounding non-tumor tissue from human NASH-HCC livers, there were strong correlations between TAZ, NOX2, and oxidative DNA damage. CONCLUSIONS: TAZ in pre-tumor NASH-hepatocytes, via induction of Cybb and NOX2-mediated DNA damage, contributes to subsequent HCC tumor development. These findings illustrate how NASH provides a unique window into the early molecular events that can lead to tumor formation and suggest that NASH therapies targeting TAZ might also prevent NASH-HCC. LAY SUMMARY: Non-alcoholic steatohepatitis (NASH) is emerging as the leading cause of a type of liver cancer called hepatocellular carcinoma (HCC), but molecular events in pre-tumor NASH hepatocytes leading to HCC remain largely unknown. Our study shows that a protein called TAZ in pre-tumor NASH-hepatocytes promotes damage to the DNA of hepatocytes and thereby contributes to eventual HCC. This study reveals a very early event in HCC that is induced in pre-tumor NASH, and the findings suggest that NASH therapies targeting TAZ might also prevent NASH-HCC.


Subject(s)
Adaptor Proteins, Signal Transducing , Carcinoma, Hepatocellular , Liver Neoplasms , NADPH Oxidase 2 , Non-alcoholic Fatty Liver Disease , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Disease Models, Animal , Hepatocytes/metabolism , Liver/pathology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Mice , Mice, Inbred C57BL , NADPH Oxidase 2/metabolism , Non-alcoholic Fatty Liver Disease/metabolism
4.
Gastroenterology ; 160(3): 781-796, 2021 02.
Article in English | MEDLINE | ID: mdl-33129844

ABSTRACT

BACKGROUND & AIMS: Immune checkpoint inhibitors have limited efficacy in many tumors. We investigated mechanisms of tumor resistance to inhibitors of programmed cell death-1 (PDCD1, also called PD-1) in mice with gastric cancer, and the role of its ligand, PD-L1. METHODS: Gastrin-deficient mice were given N-methyl-N-nitrosourea (MNU) in drinking water along with Helicobacter felis to induce gastric tumor formation; we also performed studies with H/K-ATPase-hIL1B mice, which develop spontaneous gastric tumors at the antral-corpus junction and have parietal cells that constitutively secrete interleukin 1B. Mice were given injections of an antibody against PD-1 or an isotype control before tumors developed, or anti-PD-1 and 5-fluorouracil and oxaliplatin, or an antibody against lymphocyte antigen 6 complex locus G (also called Gr-1), which depletes myeloid-derived suppressor cells [MDSCs]), after tumors developed. We generated knock-in mice that express PD-L1 specifically in the gastric epithelium or myeloid lineage. RESULTS: When given to gastrin-deficient mice before tumors grew, anti-PD-1 significantly reduced tumor size and increased tumor infiltration by T cells. However, anti-PD-1 alone did not have significant effects on established tumors in these mice. Neither early nor late anti-PD-1 administration reduced tumor growth in the presence of MDSCs in H/K-ATPase-hIL-1Ɵ mice. The combination of 5-fluorouracil and oxaliplatin reduced MDSCs, increased numbers of intra-tumor CD8+ T cells, and increased the response of tumors to anti-PD-1; however, this resulted in increased tumor expression of PD-L1. Expression of PD-L1 by tumor or immune cells increased gastric tumorigenesis in mice given MNU. Mice with gastric epithelial cells that expressed PD-L1 did not develop spontaneous tumors, but they developed more and larger tumors after administration of MNU and H felis, with accumulation of MDSCs. CONCLUSIONS: In mouse models of gastric cancer, 5-fluorouracil and oxaliplatin reduce numbers of MDSCs to increase the effects of anti-PD-1, which promotes tumor infiltration by CD8+ T cells. However, these chemotherapeutic agents also induce expression of PD-L1 by tumor cells. Expression of PD-L1 by gastric epithelial cells increases tumorigenesis in response to MNU and H felis, and accumulation of MDSCs, which promote tumor progression. The timing and site of PD-L1 expression is therefore important in gastric tumorigenesis and should be considered in design of therapeutic regimens.


Subject(s)
Helicobacter Infections/immunology , Myeloid-Derived Suppressor Cells/immunology , Neoplasms, Experimental/immunology , Programmed Cell Death 1 Receptor/metabolism , Stomach Neoplasms/immunology , Administration, Oral , Animals , Carcinogenesis/chemically induced , Carcinogenesis/drug effects , Carcinogenesis/genetics , Carcinogenesis/immunology , Gastric Mucosa/immunology , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Gastrins/genetics , Helicobacter Infections/chemically induced , Helicobacter Infections/genetics , Helicobacter Infections/microbiology , Helicobacter felis/immunology , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Methylnitrosourea/administration & dosage , Mice , Mice, Knockout , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/microbiology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/immunology , Stomach Neoplasms/chemically induced , Stomach Neoplasms/drug therapy , Stomach Neoplasms/microbiology , Tumor Microenvironment/immunology
5.
Mod Pathol ; 33(11): 2147-2155, 2020 11.
Article in English | MEDLINE | ID: mdl-32792598

ABSTRACT

The novel coronavirus SARS-CoV-2 (coronavirus disease 19, or COVID-19) primarily causes pulmonary injury, but has been implicated to cause hepatic injury, both by serum markers and histologic evaluation. The histologic pattern of injury has not been completely described. Studies quantifying viral load in the liver are lacking. Here we report the clinical and histologic findings related to the liver in 40 patients who died of complications of COVID-19. A subset of liver tissue blocks were subjected to polymerase chain reaction (PCR) for viral ribonucleic acid (RNA). Peak levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) were elevated; median ALT peak 68 U/l (normal up to 46 U/l) and median AST peak 102 U/l (normal up to 37 U/l). Macrovesicular steatosis was the most common finding, involving 30 patients (75%). Mild lobular necroinflammation and portal inflammation were present in 20 cases each (50%). Vascular pathology, including sinusoidal microthrombi, was infrequent, seen in six cases (15%). PCR of liver tissue was positive in 11 of 20 patients tested (55%). In conclusion, we found patients dying of COVID-19 had biochemical evidence of hepatitis (of variable severity) and demonstrated histologic findings of macrovesicular steatosis and mild acute hepatitis (lobular necroinflammation) and mild portal inflammation. We also identified viral RNA in a sizeable subset of liver tissue samples.


Subject(s)
Coronavirus Infections/complications , Liver Diseases/pathology , Liver Diseases/virology , Pneumonia, Viral/complications , Aged , Aged, 80 and over , Betacoronavirus , COVID-19 , Female , Humans , Male , Pandemics , SARS-CoV-2
6.
Gastroenterology ; 150(1): 218-228.e12, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26408346

ABSTRACT

BACKGROUND & AIMS: Activin, a member of the transforming growth factor-Ɵ (TGFB) family, might be involved in pancreaticĀ tumorigenesis, similar to other members of the TGFB family.Ā Human pancreatic ductal adenocarcinomas contain somatic mutations in the activin A receptor type IB (ACVR1B) gene, indicating that ACVR1B could be a suppressor of pancreatic tumorigenesis. METHODS: We disrupted Acvr1b specifically in pancreata of mice (Acvr1b(flox/flox);Pdx1-Cre mice) and crossed them with LSL-KRAS(G12D) mice, which express an activated form of KRAS and develop spontaneous pancreatic tumors. The resulting Acvr1b(flox/flox);LSL-KRAS(G12D);Pdx1-Cre mice were monitored; pancreatic tissues were collected and analyzed by histology and immunohistochemical analyses. We also analyzed p16(flox/flox);LSL-Kras(G12D);Pdx1-Cre mice and Cre-negative littermates (controls). Genomic DNA, total RNA, andĀ protein were isolated from mouse tissues and primary pancreatic tumor cell lines and analyzed by reverse-transcription polymerase chain reaction, sequencing, and immunoblot analyses. Human intraductal papillary mucinous neoplasm (IPMN) specimens were analyzed by immunohistochemistry. RESULTS: Loss of ACVR1B from pancreata of mice increased the proliferation of pancreatic epithelial cells, led toĀ formation of acinar to ductal metaplasia, and induced focalĀ inflammatory changes compared with control mice. DisruptionĀ of Acvr1b in LSL-KRAS(G12D);Pdx1-Cre mice accelerated theĀ growth of pancreatic IPMNs compared with LSL-KRAS(G12D);Pdx1-Cre mice, but did not alter growth of pancreatic intraepithelial neoplasias. We associated perinuclear localization of the activated NOTCH4 intracellular domain to the apical cytoplasm of neoplastic cells with the expansion of IPMN lesions in Acvr1b(flox/flox);LSL-KRAS(G12D);Pdx1-Cre mice. Loss of the gene that encodes p16 (Cdkn2a) was required for progression of IPMNs to pancreatic ductal adenocarcinomas in Acvr1b(flox/flox);LSL-Kras(G12D);Pdx1-Cre mice. We also observed progressive loss of p16 in human IPMNs of increasing grades. CONCLUSIONS: Loss of ACVR1B accelerates growth of mutant KRAS-induced pancreatic IPMNs in mice; this process appears to involve NOTCH4 and loss of p16. ACVR1B suppresses early stages of pancreatic tumorigenesis; the activin signaling pathway therefore might be a therapeutic target for pancreatic cancer.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Genetic Predisposition to Disease , Membrane Proteins/genetics , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Adenocarcinoma, Mucinous/genetics , Adenocarcinoma, Mucinous/mortality , Adenocarcinoma, Mucinous/pathology , Animals , Carcinogenesis/genetics , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/pathology , Disease Models, Animal , Disease Progression , Gene Deletion , Genes, Tumor Suppressor , Humans , Immunohistochemistry , Male , Mice , Mice, Knockout , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Random Allocation , Real-Time Polymerase Chain Reaction , Signal Transduction , Survival Rate
7.
Mod Pathol ; 30(2): 286-296, 2017 02.
Article in English | MEDLINE | ID: mdl-27739435

ABSTRACT

Anorectal melanoma is a rare disease that carries a poor prognosis. To date, limited genetic analyses confirmed KIT mutations as a recurrent genetic event similar to other mucosal melanomas, occurring in up to 30% of anorectal melanomas. Importantly, a subset of tumors harboring activating KIT mutations have been found to respond to c-Kit inhibitor-based therapy, with improved patient survival at advanced tumor stages. We performed comprehensive targeted exon sequencing analysis of 467 cancer-related genes in a larger series of 15 anorectal melanomas, focusing on potentially actionable variants based on gain- and loss-of-function mutations. We report the identification of oncogenic driver events in the majority (93%) of anorectal melanomas. These included variants in canonical MAPK pathway effectors rarely observed in cutaneous melanomas (including an HRAS mutation, as well as a BRAF mutation resulting in duplication of threonine 599), and recurrent mutations in the tumor suppressor NF1 in 20% of cases, which represented the second-most frequently mutated gene after KIT in our series. Furthermore, we identify SF3B1 mutations as a recurrent genetic event in mucosal melanomas. Our findings provide an insight into the genetic diversity of anorectal melanomas, and suggest significant potential for alternative targeted therapeutics in addition to c-Kit inhibitors for this melanoma subtype.


Subject(s)
Anus Neoplasms/genetics , Melanoma/genetics , Phosphoproteins/genetics , Proto-Oncogene Proteins B-raf/genetics , RNA Splicing Factors/genetics , Rectal Neoplasms/genetics , Aged , Aged, 80 and over , Anus Neoplasms/pathology , Exons , Female , Humans , Male , Melanoma/pathology , Middle Aged , Mutation , Neurofibromin 1/genetics , Rectal Neoplasms/pathology , Signal Transduction/genetics
8.
J Magn Reson Imaging ; 46(2): 393-402, 2017 08.
Article in English | MEDLINE | ID: mdl-28152252

ABSTRACT

PURPOSE: To assess the relationship between diffusion-weighted imaging (DWI) and intravoxel incoherent motion (IVIM)-derived quantitative parameters (apparent diffusion coefficient [ADC], perfusion fraction [f], Dslow , diffusion coefficient [D], and Dfast , pseudodiffusion coefficient [D*]) and histopathology in pancreatic adenocarcinoma (PAC). MATERIALS AND METHODS: Subjects with suspected surgically resectable PAC were prospectively enrolled in this Health Insurance Portability and Accountability Act (HIPAA)-compliant, Institutional Review Board-approved study. Imaging was performed at 1.5T with a respiratory-triggered echo planar DWI sequence using 10 b values. Two readers drew regions of interest (ROIs) over the tumor and adjacent nontumoral tissue. Monoexponential and biexponential fits were used to derive ADC2b , ADCall , f, D, and D*, which were compared to quantitative histopathology of fibrosis, mean vascular density, and cellularity. Two biexponential IVIM models were investigated and compared: 1) nonlinear least-square fitting based on the Levenberg-Marquardt algorithm, and 2) linear fit using a fixed D* (20 mm2 /s). Statistical analysis included Student's t-test, Pearson correlation (P < 0.05 was considered significant), intraclass correlation, and coefficients of variance. RESULTS: Twenty subjects with PAC were included in the final cohort. Negative correlation between D and fibrosis (Reader 2: r = -0.57 P = 0.01; pooled P = -0.46, P = 0.04) was observed with a trend toward positive correlation between f and fibrosis (r = 0.44, P = 0.05). ADC2b was significantly lower in PAC with dense fibrosis than with loose fibrosis ADC2b (P = 0.03). Inter- and intrareader agreement was excellent for ADC, D, and f. CONCLUSION: In PAC, D negatively correlates with fibrosis, with a trend toward positive correlation with f suggesting both perfusion and diffusion effects contribute to stromal desmoplasia. ADC2b is significantly lower in tumors with dense fibrosis and may serve as a biomarker of fibrosis architecture. LEVEL OF EVIDENCE: 1 Technical Efficacy: Stage 2 J. MAGN. RESON. IMAGING 2017;46:393-402.


Subject(s)
Adenocarcinoma/diagnostic imaging , Diffusion Magnetic Resonance Imaging , Pancreatic Neoplasms/diagnostic imaging , Adenocarcinoma/physiopathology , Adult , Aged , Algorithms , Biomarkers , Female , Fibrosis , Healthy Volunteers , Humans , Image Interpretation, Computer-Assisted , Image Processing, Computer-Assisted , Linear Models , Male , Middle Aged , Motion , Pancreatic Neoplasms/physiopathology , Prospective Studies , Reproducibility of Results , Young Adult , Pancreatic Neoplasms
9.
Histopathology ; 66(4): 598-602, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25378077

ABSTRACT

AIMS: Bile salt export pump (BSEP) is a transporter expressed exclusively at hepatic canaliculi and drives bile-salt efflux. Minimal data exist about BSEP expression in tumours. We hypothesized that BSEP immunohistochemistry would be specific for hepatocellular carcinoma (HCC). METHODS AND RESULTS: Tissue microarrays, including 48 HCC, 41 cholangiocarcinomas and 24 metastatic tumours in liver, were immunostained for BSEP. Expression was compared with common markers of hepatocytic differentiation including CD10, hepatocyte paraffin-1 antigen (HepPar-1), carcinoembryonic antigen, arginase-1 (ARG) and glypican-3 (GPC-3). BSEP expression was assessed in normal tissues. Special attention was given to adrenal gland (normal and neoplasia). BSEP was easy to interpret and showed no background staining. Canalicular expression was seen in all normal livers, but not in other normal tissue. BSEP was 90% sensitive and 100% specific for HCC (canalicular in 33 of 43 positive cases). The sensitivity of ARG was slightly higher, but specificity was slightly lower (94% for both). HepPar-1 was 90% sensitive and 97% specific. CD10, polyclonal carcinoembryonic antigen (pCEA) and GPC-3 all had lower sensitivity (74, 81 and 54%, respectively). CONCLUSIONS: In malignant tumours in the liver, BSEP marking was 100% specific and 90% sensitive for HCC. The specificity of BSEP for HCC obviates the need to identify a 'canalicular' pattern, which can limit the utility of other canalicular markers.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Bile Duct Neoplasms/diagnosis , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/diagnosis , Cholangiocarcinoma/diagnosis , Liver Neoplasms/diagnosis , ATP Binding Cassette Transporter, Subfamily B, Member 11 , Aged , Bile Duct Neoplasms/metabolism , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic/metabolism , Bile Ducts, Intrahepatic/pathology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/pathology , Diagnosis, Differential , Female , Humans , Immunohistochemistry , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Middle Aged , Sensitivity and Specificity
11.
Virchows Arch ; 484(1): 61-69, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37924345

ABSTRACT

Hemophagocytic lymphohistiocytosis (HLH) is a rare disease with high mortality. Liver involvement is common (based on elevated liver function tests) with most patients demonstrating acute hepatitis. Liver biopsies are frequently obtained in the setting of suspected HLH for the purpose of identification of erythrophagocytosis, and if present, this finding is thought to suggest or support the diagnosis of HLH. However, there are problems with this approach; in particular, we do not know whether this finding is reproducible or whether it is specific to HLH. Therefore, we conducted a multi-institutional study in which experienced liver pathologists reviewed images taken from liver biopsies from patients with normal liver, acute hepatitis, possible HLH, and clinical HLH to determine if there was agreement about the presence or absence of erythrophagocytosis, and to ascertain whether the finding corresponds to a clinical diagnosis of HLH. Twelve liver pathologists reviewed 141 images in isolation (i.e., no clinical information or diagnosis provided). These came from 32 patients (five normal, 17 acute hepatitis, six HLH, four possible HLH). The pathologists classified each image as negative, equivocal, or positive for erythrophagocytosis. Kappa was .08 (no agreement) for case-level and 0.1 for image-level (1.4% agreement, based on two images which were universally considered negative). There was no difference in the proportion of pathologists who diagnosed erythrophagocytosis among those with different diagnoses at case or image-level (p = 0.82 and p = 0.82, respectively). Thus, erythrophagocytosis is an entirely unreliable histologic parameter in liver, as it is irreproducible and not demonstrably associated with a clinical disease (namely, HLH). Unless and until more reliable guidelines can be established, pathologists should refrain from commenting on the presence or absence of erythrophagocytosis in liver biopsy.


Subject(s)
Hepatitis , Lymphohistiocytosis, Hemophagocytic , Humans , Lymphohistiocytosis, Hemophagocytic/diagnosis , Lymphohistiocytosis, Hemophagocytic/complications , Lymphohistiocytosis, Hemophagocytic/pathology , Acute Disease , Biopsy
12.
ACG Case Rep J ; 10(2): e01002, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36891182

ABSTRACT

The 2022 Mpox outbreak has caused public health concerns worldwide. Mpox infection often manifests as papular skin lesions; other systemic complications have also been reported. We present the case of a 35-year-old man with HIV who presented with rectal pain and hematochezia and was found to have severe ulceration and exudate on sigmoidoscopy consistent with Mpox proctitis.

13.
Oncoimmunology ; 11(1): 2066767, 2022.
Article in English | MEDLINE | ID: mdl-35558160

ABSTRACT

Patients with pancreatic ductal adenocarcinoma (PDAC) have a grim prognosis despite complete surgical resection and intense systemic therapies. While immunotherapies have been beneficial with many different types of solid tumors, they have almost uniformly failed in the treatment of PDAC. Understanding how therapies affect the tumor immune microenvironment (TIME) can provide insights for the development of strategies to treat PDAC. We used quantitative multiplexed immunofluorescence (qmIF) quantitative spatial analysis (qSA), and immunogenomic (IG) analysis to analyze formalin-fixed paraffin embedded (FFPE) primary tumor specimens from 44 patients with PDAC including 18 treated with neoadjuvant chemoradiation (CRT) and 26 patients receiving no treatment (NT) and compared them with tissues from 40 treatment-naĆÆve melanoma patients. We find that relative to NT tumors, CD3+ T cell infiltration was increased in CRT treated tumors (pĀ =Ā .0006), including increases in CD3+CD8+ cytotoxic T cells (CTLs, p =Ā .0079), CD3+CD4+FOXP3- T helper cells (Th, p =Ā .0010), and CD3+CD4+FOXP3+ regulatory T cells (Tregs, p =Ā .0089) with no difference in CD68+ macrophages. IG analysis from micro-dissected tissues indicated overexpression of genes involved in antigen presentation, T cell activation, and inflammation in CRT treated tumors. Among treated patients, a higher ratio of Tregs to total T cells was associated with shorter survival time (pĀ =Ā .0121). Despite comparable levels of infiltrating T cells in CRT PDACs to melanoma, PDACs displayed distinct spatial profiles with less T cell clustering as defined by nearest neighbor analysis (pĀ <Ā .001). These findings demonstrate that, while CRT can achieve high T cell densities in PDAC compared to melanoma, phenotype and spatial organization of T cells may limit benefit of T cell infiltration in this immunotherapy-resistant tumor.


Subject(s)
Carcinoma, Pancreatic Ductal , Melanoma , Pancreatic Neoplasms , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/therapy , Forkhead Transcription Factors , Humans , Melanoma/therapy , Neoadjuvant Therapy , Pancreatic Neoplasms/therapy , Tumor Microenvironment , Pancreatic Neoplasms
14.
Am J Clin Pathol ; 155(1): 124-132, 2021 01 04.
Article in English | MEDLINE | ID: mdl-32914850

ABSTRACT

OBJECTIVES: Special AT-rich binding protein 2 (SATB2) immunohistochemistry (IHC) has high sensitivity and specificity for colorectal adenocarcinoma (CRC), but data on its expression in specific subsets of pulmonary, gastric, small bowel, and pancreatobiliary adenocarcinomas (ADCAs) are relatively limited or discordant. We assessed SATB2 expression in a large cohort of ADCAs from these sites to determine its reliability in distinguishing CRC from them. METHODS: SATB2 IHC was performed on 335 neoplasms, including 40 lung ADCAs, 165 pancreatobiliary neoplasms (34 intraductal papillary mucinous neoplasms [IPMNs], 19 pancreatic ADCAs, 112 cholangiocarcinomas [CCs]), and 35 gastric, 13 small bowel, 36 ampullary (AMP), and 46 CRC ADCAs. The cases were evaluated for positivity (defined as ≥5% nuclear staining), and an H-score was calculated based on the percentage of SATB2+ cells and staining intensity. Analysis was performed to determine the optimal H-score threshold to separate CRC and non-CRC. RESULTS: SATB2 was positive in 3% of lung, 2% of CC, 17% of gastric, 38% of small bowel, and 6% of AMP ADCAs. All pancreatic ADCA/IPMNs were negative, and 87% CRCs were positive. CONCLUSIONS: SATB2 is not entirely specific for colorectal origin and can be expressed in a subset of gastrointestinal ADCAs. It is most useful in the differential of CRC vs lung and pancreatobiliary ADCAs.


Subject(s)
Adenocarcinoma/metabolism , Bile Duct Neoplasms/metabolism , Cholangiocarcinoma/metabolism , Gastrointestinal Neoplasms/metabolism , Lung Neoplasms/metabolism , Matrix Attachment Region Binding Proteins/metabolism , Pancreatic Neoplasms/metabolism , Transcription Factors/metabolism , Adenocarcinoma/pathology , Bile Duct Neoplasms/pathology , Cholangiocarcinoma/pathology , Gastrointestinal Neoplasms/pathology , Humans , Lung Neoplasms/pathology , Pancreatic Neoplasms/pathology
15.
J Clin Pathol ; 74(2): 98-101, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32471888

ABSTRACT

AIMS: In situ hybridisation (ISH) for albumin mRNA is a sensitive marker of primary liver tumours in adults. However, paediatric tumours, such as hepatoblastoma (HB) and fibrolamellar hepatocellular carcinoma (FLC), have not been tested thoroughly and may require ancillary tests to diagnose with confidence. We aim to determine if albumin ISH is useful in the pathological evaluation of these malignancies and to compare it to commonly used immunohistochemical markers HepPar 1 (HEPA) and arginase-1 (ARG). METHODS: Tissue microarrays of 26 HB and 10 FLC were constructed. Controls included 4 embryonal undifferentiated sarcomas of the liver, 51 neuroblastomas and 64 Wilms tumours. We evaluated a commercially available RNA ISH to detect albumin mRNA. Immunohistochemistry for HEPA and ARG was performed in the usual fashion. RESULTS: Twenty-six of 26 HB showed positive staining by albumin ISH including 14 fetal, 8 embryonal and 4 mixed variants. All 10 FLC were diffusely positive. The sensitivity and specificity of albumin ISH were 100% for HB and FLC. ARG had 100% sensitivity and specificity for HB (26 of 26 cases) and FLC (9 of 9). HEPA stained 22 of 26 HB (85% sensitivity, 99.2% specificity) and 7 of 9 FLC (78% sensitivity, 99.1% specificity). CONCLUSION: Albumin RNA ISH is a useful test to determine hepatocytic origin in HB and FLC. ARG was equally sensitive and easy to interpret, while HEPA was inferior to both in HB and FLC.


Subject(s)
Albumins/analysis , Biomarkers, Tumor/analysis , In Situ Hybridization/methods , Liver Neoplasms/diagnosis , RNA, Messenger/analysis , Child , Child, Preschool , Female , Humans , Immunohistochemistry , Infant , Male , Sensitivity and Specificity
16.
Oxf Med Case Reports ; 2021(11): omab113, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34858627

ABSTRACT

Wild-type ATTR cardiac amyloidosis (ATTRwt-CA) is not as rare as previously thought to be. Patients with infiltrative cardiac amyloidosis often present with right-sided heart failure (HF) symptomatology. Clinically significant liver disease and cirrhosis has not been reported in ATTRwt-CA. We present two cases of ATTRwt-CA with right-sided HF and abnormal liver function tests initially thought to be secondary to congestive hepatopathy but found to have rare and unrelated liver disease. These cases highlight the importance of developing a broad differential diagnosis and leveraging a multidisciplinary team approach in evaluating patients for unusual causes of cirrhosis/other chronic liver diseases when ATTR cardiac amyloidosis patients present with congestive hepatopathy.

17.
JCI Insight ; 6(9)2021 05 10.
Article in English | MEDLINE | ID: mdl-33755595

ABSTRACT

Most colorectal cancers (CRCs) are moderately differentiated or well differentiated, a status that is preserved even in metastatic tumors. However, the molecular mechanisms underlying CRC differentiation remain to be elucidated. Herein, we unravel a potentially novel posttranscriptional regulatory mechanism via a LIN28B/CDX2 signaling axis that plays a critical role in mediating CRC differentiation. Owing to a large number of mRNA targets, the mRNA-binding protein LIN28B has diverse functions in development, metabolism, tissue regeneration, and tumorigenesis. Our RNA-binding protein IP (RIP) assay revealed that LIN28B directly binds CDX2 mRNA, which is a pivotal homeobox transcription factor in normal intestinal epithelial cell identity and differentiation. Furthermore, LIN28B overexpression resulted in enhanced CDX2 expression to promote differentiation in subcutaneous xenograft tumors generated from CRC cells and metastatic tumor colonization through mesenchymal-epithelial transition in CRC liver metastasis mouse models. A ChIP sequence for CDX2 identified α-methylacyl-CoA racemase (AMACR) as a potentially novel transcriptional target of CDX2 in the context of LIN28B overexpression. We also found that AMACR enhanced intestinal alkaline phosphatase activity, which is known as a key component of intestinal differentiation, through the upregulation of butyric acid. Overall, we demonstrated that LIN28B promotes CRC differentiation through the CDX2/AMACR axis.


Subject(s)
Adenocarcinoma/genetics , CDX2 Transcription Factor/metabolism , Cell Differentiation/genetics , Colorectal Neoplasms/genetics , RNA-Binding Proteins/genetics , Racemases and Epimerases/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/secondary , Animals , Caco-2 Cells , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Mice , Mice, Transgenic , Neoplasm Transplantation , RNA-Binding Proteins/metabolism
18.
Cancer Cell ; 39(6): 866-882.e11, 2021 06 14.
Article in English | MEDLINE | ID: mdl-33930309

ABSTRACT

Cancer-associated fibroblasts (CAF) are a poorly characterized cell population in the context of liver cancer. Our study investigates CAF functions in intrahepatic cholangiocarcinoma (ICC), a highly desmoplastic liver tumor. Genetic tracing, single-cell RNA sequencing, and ligand-receptor analyses uncovered hepatic stellate cells (HSC) as the main source of CAF and HSC-derived CAF as the dominant population interacting with tumor cells. In mice, CAF promotes ICC progression, as revealed by HSC-selective CAF depletion. In patients, a high panCAF signature is associated with decreased survival and increased recurrence. Single-cell RNA sequencing segregates CAF into inflammatory and growth factor-enriched (iCAF) and myofibroblastic (myCAF) subpopulations, displaying distinct ligand-receptor interactions. myCAF-expressed hyaluronan synthase 2, but not type I collagen, promotes ICC. iCAF-expressed hepatocyte growth factor enhances ICC growth via tumor-expressed MET, thus directly linking CAF to tumor cells. In summary, our data demonstrate promotion of desmoplastic ICC growth by therapeutically targetable CAF subtype-specific mediators, but not by type I collagen.


Subject(s)
Bile Duct Neoplasms/pathology , Cancer-Associated Fibroblasts/pathology , Cholangiocarcinoma/pathology , Aged , Animals , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/metabolism , Bile Ducts, Intrahepatic/pathology , Cancer-Associated Fibroblasts/metabolism , Cholangiocarcinoma/genetics , Cholangiocarcinoma/metabolism , Collagen Type I/metabolism , Female , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/pathology , Hepatocyte Growth Factor/metabolism , Humans , Hyaluronan Synthases/genetics , Hyaluronan Synthases/metabolism , Hyaluronic Acid/metabolism , Male , Mice, Transgenic , Middle Aged , Proto-Oncogene Proteins c-met/metabolism , Tumor Microenvironment
19.
Cancer Cytopathol ; 127(12): 771-777, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31756042

ABSTRACT

BACKGROUND: Cell blocks are being used more frequently in cytology for ancillary testing, including molecular diagnostics. There are several different methods of processing cell blocks, with plasma-thrombin being one of the most common. Plasma is a blood-derived product and may be a source of DNA. The aim of this study was to determine whether the plasma used for the plasma-thrombin cell block method has amplifiable DNA that may potentially interfere with molecular testing results. METHODS: Expired bags of fresh frozen plasma were collected from a blood bank. From each sample, DNA was extracted from a 1-mL aliquot with the QIAsymphony MIDI kit (Qiagen). The concentration of DNA was measured on a NanoDrop instrument. The amplifiable DNA quality was assessed by polymerase chain reaction (PCR) with primers to generate amplicons of various sizes. Characterization was performed with the AmpFLSTR Identifiler Plus PCR kit with capillary electrophoresis. RESULTS: Twenty samples from 20 bags were collected. All samples showed amplifiable DNA despite low DNA concentrations in a few cases. PCR amplification revealed the presence of high-quality amplifiable DNA (up to 600 base pairs). DNA was amplified at the 16 loci interrogated in all samples tested with the AmpFLSTR Identifiler Plus PCR kit. CONCLUSIONS: The presence of genomic DNA in plasma may theoretically interfere with results of molecular testing. Particularly in clinical samples with low cellularity, the DNA in plasma may potentially either mask the presence of minute amounts of tumor-derived DNA or lead to a false-positive result.


Subject(s)
Blood Banks/statistics & numerical data , DNA Contamination , DNA Primers/administration & dosage , Plasma , Thrombin/metabolism , DNA Fingerprinting , Female , Humans , Male , Polymerase Chain Reaction/methods , Prospective Studies , Sensitivity and Specificity , Specimen Handling
20.
Cancers (Basel) ; 11(12)2019 Dec 11.
Article in English | MEDLINE | ID: mdl-31835848

ABSTRACT

The clinical management of pediatric liver tumors involves stratification into risk groups. One previously defined, high-risk group of hepatoblastomas is the small cell undifferentiated variant. In light of molecular studies showing SMARCB1 deletion in these tumors, it is now recognized that most small cell, undifferentiated liver tumors represent an aggressive unrelated tumor-the malignant rhabdoid tumor (MRT). SMARCB1 is a member of the chromatin remodeling SWI/SNF complex and encodes the INI1 protein. The histologic diagnosis of MRT is currently based on INI1 negative immunoreactivity and the presence of rhabdoid morphology. INI1-negative small cell liver tumors lacking classic rhabdoid morphology are often misclassified as small cell undifferentiated hepatoblastomas (SCUD-HB), according to the current classification. Pediatric liver tumors diagnosed between 2003-2017 as SCUD-HB (four cases) or MRT (two cases) were identified from the Columbia University Pathology Department Archives. All tumors were associated with normal or low serum alpha fetoprotein levels, and showed an absence of immunohistochemical staining of hepatocellular markers (Hep-par1, Arginase) and loss of INI1 staining. Two cases were initially diagnosed as MRT, one with prominent rhabdoid morphology, the other with predominant small cell morphology. The remaining four cases with small cell morphology were classified as SCUD-HB. Ancillary molecular studies confirmed the loss of SMARCB1, supporting the diagnosis of MRT in all cases, proving morphology an unreliable criterion. It is critical to eliminate the term INI1-negative hepatoblastoma from the current classification scheme, and classify INI1-negative tumors as MRT, particularly since high-risk HB-chemotherapy regimens are not effective for treating MRT.

SELECTION OF CITATIONS
SEARCH DETAIL