Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 286
Filter
1.
Immunity ; 56(12): 2677-2678, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-38091948

ABSTRACT

Neurodegeneration is a devastating complication of Langerhans cell histiocytosis (LCH), but it is not clear how it develops. In this issue of Immunity, Wilk et al. demonstrate that circulating BRAFV600E+ myeloid cells damage the blood-brain barrier and infiltrate the brain. Dual inhibition of the MAPK and senescence pathways can block parenchymal injury, providing a potential therapeutic avenue for histiocytic neurodegeneration.


Subject(s)
Histiocytosis, Langerhans-Cell , Monocytes , Humans , Monocytes/metabolism , Histiocytosis, Langerhans-Cell/metabolism , Brain/metabolism , Blood-Brain Barrier/metabolism , Myeloid Cells/metabolism , Proto-Oncogene Proteins B-raf/metabolism
2.
Cancer Sci ; 114(5): 1871-1881, 2023 May.
Article in English | MEDLINE | ID: mdl-36788737

ABSTRACT

Langerhans cell histiocytosis (LCH) is a rare neoplastic disorder characterized by inflammatory lesions arising from the anomalous accumulation of pathogenic CD1a+ CD207+ dendritic cells (DCs). SIRPα is a transmembrane protein highly expressed in myeloid cells such as DCs and macrophages. Here we show that SIRPα is a potential therapeutic target for LCH. We found that SIRPα is expressed in CD1a+ cells of human LCH lesions as well as in CD11c+ DCs in the spleen, liver, and lung of a mouse model of LCH (BRAFV600ECD11c mouse), in which an LCH-associated active form of human BRAF is expressed in a manner dependent on the mouse Cd11c promoter. BRAFV600ECD11c mice manifested markedly increased numbers of CD4+ T cells, regulatory T cells, and macrophages as well as of CD11c+ MHCII+ DCs in the spleen. Monotherapy with a mAb to SIRPα greatly reduced the percentage of CD11c+ MHCII+ DCs in peripheral blood, LCH-like lesion size in the liver, and the number of CD11c+ MHCII+ DCs in the spleen of the mutant mice. Moreover, this mAb promoted macrophage-mediated phagocytosis of CD11c+ DCs from BRAFV600ECD11c mice, whereas it had no effects on the viability or CCL19-dependent migration of such CD11c+ DCs or on their expression of the chemokine genes Ccl5, Ccl20, Cxcl11, and Cxcl12. Our results thus suggest that anti-SIRPα monotherapy is a promising approach to the treatment of LCH that is dependent in part on the promotion of the macrophage-mediated killing of LCH cells.


Subject(s)
Histiocytosis, Langerhans-Cell , Animals , Humans , Mice , Histiocytosis, Langerhans-Cell/drug therapy , Histiocytosis, Langerhans-Cell/genetics , Histiocytosis, Langerhans-Cell/metabolism , Spleen/metabolism
3.
Sci Immunol ; 7(78): eadd3330, 2022 12 23.
Article in English | MEDLINE | ID: mdl-36525505

ABSTRACT

Langerhans cell histiocytosis (LCH) is a potentially fatal neoplasm characterized by the aberrant differentiation of mononuclear phagocytes, driven by mitogen-activated protein kinase (MAPK) pathway activation. LCH cells may trigger destructive pathology yet remain in a precarious state finely balanced between apoptosis and survival, supported by a unique inflammatory milieu. The interactions that maintain this state are not well known and may offer targets for intervention. Here, we used single-cell RNA-seq and protein analysis to dissect LCH lesions, assessing LCH cell heterogeneity and comparing LCH cells with normal mononuclear phagocytes within lesions. We found LCH discriminatory signatures pointing to senescence and escape from tumor immune surveillance. We also uncovered two major lineages of LCH with DC2- and DC3/monocyte-like phenotypes and validated them in multiple pathological tissue sites by high-content imaging. Receptor-ligand analyses and lineage tracing in vitro revealed Notch-dependent cooperativity between DC2 and DC3/monocyte lineages during expression of the pathognomonic LCH program. Our results present a convergent dual origin model of LCH with MAPK pathway activation occurring before fate commitment to DC2 and DC3/monocyte lineages and Notch-dependent cooperativity between lineages driving the development of LCH cells.


Subject(s)
Histiocytosis, Langerhans-Cell , Neoplasms , Humans , Cell Lineage , Histiocytosis, Langerhans-Cell/metabolism , Histiocytosis, Langerhans-Cell/pathology , Cell Differentiation , Monocytes/metabolism
4.
Vnitr Lek ; 68(E-2): 11-21, 2022.
Article in English | MEDLINE | ID: mdl-36208940

ABSTRACT

Langerhans cell histiocytosis (LCH) is a rare condition with incidence in adults 1-2/1 million, wherein Langerhans cells proliferate abnormally, adversely impacting organs including most frequently bones, skin, lungs, pituitary gland, lymph nodes, gums and other organs. The LCH course varies widely among patients from a self-limiting condition, to one that progresses. But LCH only very rarely culminates in death. To aim of this text is to review all possible symptoms and manifestations of this disease.


Subject(s)
Histiocytosis, Langerhans-Cell , Adult , Histiocytosis, Langerhans-Cell/diagnosis , Histiocytosis, Langerhans-Cell/metabolism , Histiocytosis, Langerhans-Cell/therapy , Humans , Lymph Nodes/pathology , Rare Diseases
5.
J Leukoc Biol ; 112(5): 1089-1104, 2022 11.
Article in English | MEDLINE | ID: mdl-35648675

ABSTRACT

Langerhans cell histiocytosis (LCH) is an inflammatory disease characterized by abnormal dendritic cells (DCs) with hyperactive ERK signaling, called "LCH cells." Since DCs rely on ERK signaling to produce inflammatory molecules in response to pathogenic cues, we hypothesized that hyperactive ERK enhances DCs inflammatory responses. We specifically investigated TLR4-induced TNFα production in LCH cells by utilizing the BRAF-V600Efl/+ :CD11c-Cre mouse model of LCH, which hyperactivates ERK in DCs. We measured LPS-induced TNFα production both in vivo and in vitro using splenic CD11c+ cells and bone marrow-derived DCs with or without pharmacologic BRAFV600E inhibition. We observed a reversible increase in secreted TNFα and a partially reversible increase in TNFα protein per cell, despite a decrease in TLR4 signaling and Tnfa transcripts compared with controls. We examined ERK-driven, posttranscriptional mechanisms that contribute to TNFα production and secretion using biochemical and cellular assays. We identified a reversible increase in TACE activation, the enzyme required for TNFα secretion, and most strikingly, an increase in protein translation, including TNFα. Defining the translatome through polysome-bound RNA sequencing revealed up-regulated translation of the LPS-response program. These data suggest hyperactive ERK signaling utilizes multiple posttranscriptional mechanisms to amplify inflammatory responses in DCs, advancing our understanding of LCH and basic DC biology.


Subject(s)
Histiocytosis, Langerhans-Cell , Proto-Oncogene Proteins B-raf , Mice , Animals , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Lipopolysaccharides/pharmacology , Tumor Necrosis Factor-alpha/genetics , Toll-Like Receptor 4/genetics , Mutation , Histiocytosis, Langerhans-Cell/metabolism , Dendritic Cells/metabolism
6.
J Immunol ; 209(2): 270-279, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35768149

ABSTRACT

Langerhans cell histiocytosis (LCH) is a disorder characterized by an abnormal accumulation of CD207+ and CD1a+ cells in almost any tissue. Currently, there is a lack of prognostic markers to follow up patients and track disease reactivation or treatment response. Putative myeloid precursors CD207+ and CD1a+ cells were previously identified circulating in the blood. Therefore, we aim to develop a sensitive tracing method to monitor circulating CD207+ and CD1a+ cells in a drop of blood sample of patients with LCH. A total of 202 blood samples from patients with LCH and 23 controls were tested using flow cytometry. A standardized cellular score was defined by quantifying CD207+ and CD1a+ expression in monocytes and dendritic cells, based on CD11b, CD14, CD11c, and CD1c subpopulations, resulting in a unique value for each sample. The scoring system was validated by a receiver operating characteristic curve showing a reliable discriminatory capacity (area under the curve of 0.849) with a threshold value of 14, defining the presence of circulating CD207+ and CD1a+ cells. Interestingly, a fraction of patients with no evident clinical manifestation at the time of sampling also showed presence of these cells (29.6%). We also found a differential expression of CD207 and CD1a depending on the organ involvement, and a positive correlation between the cellular score and plasma inflammatory markers such as soluble CD40L, soluble IL-2Ra, and CXCL12. In conclusion, the analysis of circulating CD207 and CD1a cells in a small blood sample will allow setting a cellular score with minimal invasiveness, helping with prognostic accuracy, detecting early reactivation, and follow-up.


Subject(s)
Histiocytosis, Langerhans-Cell , Mannose-Binding Lectins , Antigens, CD/metabolism , Antigens, CD1/metabolism , Histiocytosis, Langerhans-Cell/diagnosis , Histiocytosis, Langerhans-Cell/metabolism , Humans , Langerhans Cells , Lectins, C-Type/metabolism , Mannose-Binding Lectins/metabolism
7.
Am J Ophthalmol ; 242: 36-51, 2022 10.
Article in English | MEDLINE | ID: mdl-35594918

ABSTRACT

PURPOSE: Frequent activating mutations in the mitogen-activated protein kinase (MAPK) pathway genes have been identified in histiocytoses. MAPK signaling consistently upregulates cyclin D1. The goal of this study was to determine whether cyclin D1 expression by immunohistochemistry is a useful diagnostic marker for periocular histiocytoses and to further characterize their genetic basis. DESIGN: Retrospective observational case series. METHODS: Pathology records were searched for all patients with histiocytoses diagnosed between 1995 and 2020. Eleven histiocyte-rich inflammatory lesions and 10 xanthelasma served as controls. Cyclin D1 immunohistochemistry was performed on all tissues. A subset of histiocytoses was evaluated by next-generation sequencing (NGS) and droplet digital PCR (ddPCR). RESULTS: There were 36 patients, 15 males (42%) and 21 females (58%), with histiocytoses: 9 juvenile xanthogranuloma (25%), 8 adult-onset asthma and periocular xanthogranuloma (22%), 7 Langerhans cell histiocytosis (19%), 5 Rosai-Dorfman disease (14%), 5 xanthogranuloma-not otherwise specified (14%), 1 Erdheim-Chester disease (3%), and 1 histiocytic sarcoma (3%). Moderate to strong nuclear cyclin D1 expression was present in ≥50% of lesional cells in histiocytoses (23/36, 64%), significantly more when compared to histiocyte-rich inflammatory lesions (0/11, 0%, P<.001) and xanthelasma (0/10, 0%, P<.001). Cyclin D1 was expressed in <10% of lesional cells in all 11 histiocyte-rich inflammatory lesions (P<.001) and all 10 xanthelasma lesions (P<.001). MAPK pathway gene mutations were detected in 12 of 14 (86%) histiocytoses successfully assayed by NGS and/or ddPCR. CONCLUSIONS: Our study confirms that the cyclin D1 immunohistochemical stain is a useful diagnostic marker for periocular histiocytoses, correlating with underlying mutations in MAPK pathway genes.


Subject(s)
Histiocytosis, Langerhans-Cell , Neoplasms , Adult , Cyclin D1/genetics , Dendritic Cells/metabolism , Dendritic Cells/pathology , Female , Histiocytosis, Langerhans-Cell/genetics , Histiocytosis, Langerhans-Cell/metabolism , Histiocytosis, Langerhans-Cell/pathology , Humans , Macrophages/metabolism , Macrophages/pathology , Male , Mitogen-Activated Protein Kinases , Molecular Biology , Retrospective Studies
8.
Tohoku J Exp Med ; 255(1): 27-31, 2021 09.
Article in English | MEDLINE | ID: mdl-34497165

ABSTRACT

Langerhans cell neoplasms, which include Langerhans cell histiocytosis and Langerhans cell sarcoma, are tumors that originate from dendritic cells. Langerhans cell sarcoma is defined as a high-grade neoplasm with overtly malignant cytological features and the Langerhans cell-like phenotype, and generally has a poorer prognosis and more aggressive phenotype than Langerhans cell histiocytosis. Insulin-like growth factor 2 messenger RNA-binding protein 3 (IGF2BP3 or IMP3) is an oncofetal protein that is expressed in various cancer types; its expression is often associated with a poor prognosis and aggressive phenotype. Here, we used immunohistochemistry to evaluate IGF2BP3 expression in Langerhans cell neoplasms. IGF2BP3 expression was scored as negative (< 1%) or positive (≥ 1%) by immunohistochemistry. All 4 patients with Langerhans cell sarcoma (100%) and 6 of 22 pediatric (age < 18 years) patients with Langerhans cell histiocytosis (27.3%) had positive results for IGF2BP3; however, 16 of 22 pediatric patients with Langerhans cell histiocytosis (72.7%) and all 15 adult (age ≥ 18 years) patients with Langerhans cell histiocytosis (100%) had a negative result. Among patients with Langerhans cell histiocytosis, IGF2BP3 expression was independent of sex, location, prognosis, and BRAF V600E staining results. Taken together, these results indicate that IGF2BP3 expression may be a helpful marker for distinguishing Langerhans cell sarcoma from Langerhans cell histiocytosis in adult patients.


Subject(s)
Histiocytosis, Langerhans-Cell/metabolism , Langerhans Cell Sarcoma/metabolism , RNA-Binding Proteins/metabolism , Adolescent , Adult , Biomarkers/metabolism , Biomarkers, Tumor/metabolism , Child , Child, Preschool , Diagnosis, Differential , Female , Histiocytosis, Langerhans-Cell/diagnosis , Humans , Immunohistochemistry , Infant , Infant, Newborn , Langerhans Cell Sarcoma/diagnosis , Male , Middle Aged , Young Adult
9.
Ann Diagn Pathol ; 52: 151720, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33676171

ABSTRACT

Transdifferentiation of follicular lymphoma to a Langerhans cell neoplasm is rarely reported and not well understood. Here we present a case, review the literature and discuss some of the biological underpinnings of lineage switch of B cells to histiocytes/Langerhans cells. A 31-year-old woman had follicular lymphoma (FL) and Langerhans cell sarcoma (LCS) co-localized above and below diaphragm. The FL was low-grade, had typical morphologic features, and was positive for CD10, BCL-2, and BCL-6. The LCS was cytologically atypical with necrosis and a high mitotic rate, and the immunophenotype supported Langerhans cell lineage positive for CD1a, CD207/langerin, and S-100 protein. Both tumors carried IGH-BCL2 and the LCS cells had immunophenotypic evidence of a residual B cell program, supporting the notion that these neoplasms are clonally related. The case reported is unusual because the patient was young and both diseases presented simultaneously, before any therapy. In addition, immunohistochemical analysis showed that the LCS was negative for BRAF V600E and phospho-ERK, suggesting that the LCS belongs to the known subset of Langerhans cell tumors lacking BRAF V600E and MAP2K1 mutations. Concurrent occurrence of FL and Langerhans cell neoplasm is an unusual phenomenon, with 10 cases reported previously: 4 Langerhans cell histiocytosis and 6 Langerhans cell sarcoma, including this case.


Subject(s)
Histiocytosis, Langerhans-Cell/diagnosis , Langerhans Cell Sarcoma/diagnosis , Lymphoma, Follicular/diagnosis , Neoplasms, Second Primary/pathology , Adult , Aged , B-Lymphocytes/pathology , Cell Dedifferentiation/genetics , Cell Transdifferentiation/genetics , Female , Histiocytes/pathology , Histiocytosis, Langerhans-Cell/metabolism , Histiocytosis, Langerhans-Cell/pathology , Humans , Immunohistochemistry/methods , Immunophenotyping/methods , Langerhans Cell Sarcoma/metabolism , Langerhans Cell Sarcoma/pathology , Langerhans Cells/pathology , Lymphoma, Follicular/metabolism , Lymphoma, Follicular/pathology , Male , Middle Aged , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Neoplasm Staging/methods
10.
Pathol Int ; 71(3): 191-198, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33497038

ABSTRACT

The neonatal Fc receptor (FcRn) plays a role in trafficking IgG and albumin and is thought to mediate intravenous immunoglobulin (IVIG) therapy for certain diseases. IVIG can be used for the treatment of human Langerhans cell histiocytosis (LCH); however, the mechanism remains unclear. The expression and function of FcRn protein have not been studied in LCH, though the expression of FcRn messenger RNA (mRNA) have been reported. In this report, we confirmed the expression of FcRn in 26 of 30 pathological cases (86.7%) diagnosed immunohistochemically as LCH. The expression was independent of age, gender, location, multi- or single-system, and the status of BRAFV600E immunostaining. We also confirmed the expression of FcRn mRNA and protein in the human LCH-like cell line, ELD-1. FcRn suppressed albumin consumption and growth of IVIG preparation-treated ELD-1 cells, but not of IVIG preparation-untreated or FcRn-knockdown ELD-1 cells. In addition, FITC-conjugated albumin was taken into Rab11-positive recycle vesicles in mock ELD-1 cells but not in FcRn-knockdown ELD-1 cells. IVIG preparation prolonged this status in mock ELD-1 cells. Therefore, ELD-1 recycled albumin via FcRn and albumin was not used for metabolism. Our results increase our understanding of the molecular mechanism of IVIG treatment of LCH.


Subject(s)
Histiocytosis, Langerhans-Cell , Histocompatibility Antigens Class I/metabolism , Immunoglobulins, Intravenous , Receptors, Fc/metabolism , Adolescent , Adult , Albumins/metabolism , Cell Line , Child , Child, Preschool , Female , Fluorescein-5-isothiocyanate/analogs & derivatives , Fluorescein-5-isothiocyanate/metabolism , Histiocytosis, Langerhans-Cell/drug therapy , Histiocytosis, Langerhans-Cell/metabolism , Humans , Immunoglobulins, Intravenous/metabolism , Immunoglobulins, Intravenous/pharmacology , Infant , Infant, Newborn , Male , Serum Albumin/metabolism
11.
Am J Surg Pathol ; 44(12): 1658-1665, 2020 12.
Article in English | MEDLINE | ID: mdl-32910018

ABSTRACT

Langerhans cell histiocytosis (LCH) is a rare histiocytic disorder composed of Langerhans cells admixed with reactive mononuclear and granulocytic cells, associated with prominent eosinophils. LCH is considered a neoplasm, driven in most cases by oncogenic RAS/RAF/MEK/ERK pathway mutations. The disease predominantly affects children. Urinary system involvement has rarely been reported in a multisystem disease setting. We describe 7 patients who presented with LCH occurring within (6 cases) or after (1 case) a resected clear cell (n=6) or clear cell papillary (n=1) renal cell carcinoma (RCC), identified prospectively in our routine and consultation files (2012 to 2019). The patients included 5 women and 2 men, with a median age of 54 years (range, 39 to 73 y), none with a history of LCH or LCH manifestations before the time of RCC diagnosis. The median size of the RCC was 3.5 cm (range, 1.8 to 8.3 cm). Treatment included partial (5 cases), or radical (2 cases) nephrectomy. All RCCs on gross examination showed at least focal cystic changes and were low grade (World Health Organization [WHO]/International Society of Urologic Pathologists [ISUP] grade 1 to 2). The LCH foci were detected as incidental histological finding within the resected RCC in all six cases and they were limited to few high-power fields (<2 mm) in 5 of 6 cases, but in the sixth case, they occupied almost the entire clear cell papillary RCC (2 cm nodule). No LCH manifestations were detected in the normal kidney or in perinephric fat. The seventh patient developed LCH within inguinal deep soft tissue followed by systemic manifestations 6 years after clear cell RCC. Langerhans cell immunophenotype was supported by the reactivity for S-100, CD1a, and langerin and by the negative pankeratin. Successful pyrosequencing of microdissected LCH DNA revealed the V600E BRAF mutation in all 6 cases of LCH within RCC. To our knowledge, only 3 similar cases were published since 1980; the only case tested for BRAF mutation showed wild-type BRAF. This is the first study analyzing the morphologic and genetic features of a cohort of LCH associated with RCC. In our experience, these cases may be underrecognized in practice, or may erroneously be diagnosed as RCC dedifferentiation or high-grade sarcomatoid transformation. Finally, the detection of BRAF mutation further confirms that LCH in this setting is indeed a neoplasm, rather than a reactive lesion.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Histiocytosis, Langerhans-Cell/genetics , Histiocytosis, Langerhans-Cell/pathology , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Mutation , Proto-Oncogene Proteins B-raf/genetics , Adult , Aged , Biomarkers, Tumor/analysis , Carcinoma, Renal Cell/chemistry , Carcinoma, Renal Cell/surgery , Female , Histiocytosis, Langerhans-Cell/metabolism , Histiocytosis, Langerhans-Cell/surgery , Humans , Kidney Neoplasms/chemistry , Kidney Neoplasms/surgery , Male , Middle Aged , Neoplasm Grading , Nephrectomy , Prospective Studies
12.
Am J Med Genet A ; 182(11): 2746-2750, 2020 11.
Article in English | MEDLINE | ID: mdl-32945094

ABSTRACT

Pitt-Hopkins syndrome (PTHS, MIM #610954) is a rare neurodevelopmental disease characterized by the association of intellectual disability, characteristic facial gestalt and episodes of abnormal and irregular breathing. PTHS is due to heterozygous loss-of-function variants in the TCF4 gene (transcription factor 4, MIM #602272) encoding for a basic helix-loop-helix transcription factor. TCF4 is highly expressed during early development of the nervous system, and it is involved in cellular differentiation and proliferation. Since the first clinical description in 1978, less than 200 PTHS patients have been described. A comprehensive phenotype, especially regarding cancer predisposition, is not yet well defined. We report the case of a 7-year-old boy affected by PTHS with a 4-week history of progressive swelling of the frontal bones diagnosed with Langerhans cell histiocytosis.


Subject(s)
Histiocytosis, Langerhans-Cell/pathology , Hyperventilation/complications , Intellectual Disability/complications , Mutation , Transcription Factor 4/genetics , Child , Facies , Histiocytosis, Langerhans-Cell/etiology , Histiocytosis, Langerhans-Cell/metabolism , Humans , Male , Phenotype
14.
Scand J Immunol ; 92(6): e12950, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32738155

ABSTRACT

Langerhans cell histiocytosis (LCH) is an inflammatory myeloid neoplasia commonly affecting children with frequent somatic mutations in MAPK pathway genes including BRAFV600E and MAP2K1. Some studies suggest that LCH cells can recruit and modulate inflammatory cells, which could provide reciprocal survival signals. To characterize the immune profile of infiltrating inflammatory cells, and to clarify their participation in LCH pathogenesis, a detailed immunohistochemical analysis was performed. Fifteen (10 children, 5 adults) LCH cases were assessed through macrophage (CD68 and CD163), mature dendritic cell (mDC; CD83 and CD208), regulatory T cell (Treg; CD4, CD25 and FOXP3) and cytotoxic lymphocyte (CL; CD56, CD57, perforin and granzyme B) immunomarkers. Moreover, lymphocytic and LCH markers were also analysed. All cases were S100, CD1a, CD207 and CD4-positive. Bcl-2 and cyclin D1 expression was observed in 13 of 15 cases. In the immune microenvironment, M2-polarized macrophages and Tregs were the predominant cell populations, followed by significantly (P < .005) smaller levels of mDCs and CLs. Additionally, the number of CD3 + cells was significantly higher than that of CD20 + cells. In the CD3 + cell population, there were a significantly higher number of CD4 + cells than CD8 + cells. While there were no differences when comparing the paediatric and adult populations, FOXP3 + cells were significantly higher in patients with multisystem involvement and treated with chemotherapy, than single-site cases and those without chemotherapy. Our results suggest that M2-polarized macrophages and Treg infiltration can promote LCH development and survival, probably through pro-tumoral, immunosuppressive and/or cytokine-mediated mechanisms. This work highlights the need for further exploration of immune-targeted therapy for LCH.


Subject(s)
Histiocytosis, Langerhans-Cell/metabolism , Langerhans Cells/physiology , Macrophages/metabolism , T-Lymphocytes, Regulatory/metabolism , Adult , Antigens, CD/metabolism , Cell Differentiation , Cellular Microenvironment , Child , Child, Preschool , Cytokines/metabolism , Dendritic Cells/metabolism , Female , Forkhead Transcription Factors/metabolism , Humans , Immunohistochemistry/methods , Infant , Macrophages/immunology , Male , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Regulatory/immunology , Th2 Cells/immunology
15.
Blood ; 136(19): 2188-2199, 2020 11 05.
Article in English | MEDLINE | ID: mdl-32750121

ABSTRACT

Langerhans cell histiocytosis (LCH) is a myeloid neoplasia, driven by sporadic activating mutations in the MAPK pathway. The misguided myeloid dendritic cell (DC) model proposes that high-risk, multisystem, risk-organ-positive (MS-RO+) LCH results from driver mutation in a bone marrow (BM)-resident multipotent hematopoietic progenitor, while low-risk, MS-RO- and single-system LCH would result from driver mutation in a circulating or tissue-resident, DC-committed precursor. We have examined the CD34+c-Kit+Flt3+ myeloid progenitor population as potential mutation carrier in all LCH disease manifestations. This population contains oligopotent progenitors of monocytes (Mo's)/macrophages (MΦs), osteoclasts (OCs), and DCs. CD34+c-Kit+Flt3+ cells from BM of MS-RO+ LCH patients produced Langerhans cell (LC)-like cells in vitro. Both LC-like and DC offspring from this progenitor carried the BRAF mutation, confirming their common origin. In both high- and low-risk LCH patients, CD34+c-Kit+Flt3+ progenitor frequency in blood was higher than in healthy donors. In one MS-RO+ LCH patient, CD34+c-Kit+Flt3+ cell frequency in blood and its BRAF-mutated offspring reported response to chemotherapy. CD34+c-Kit+Flt3+ progenitors from blood of both high- and low-risk LCH patients gave rise to DCs and LC-like cells in vitro, but the driver mutation was not easily detectable, likely due to low frequency of mutated progenitors. Mutant BRAF alleles were found in Mo's /MΦs, DCs, LC-like cells, and/or OC-like cells in lesions and/or Mo and DCs in blood of multiple low-risk patients. We therefore hypothesize that in both high- and low-risk LCH, the driver mutation is present in a BM-resident myeloid progenitor that can be mobilized to the blood.


Subject(s)
Bone Marrow/pathology , Cell Differentiation , Dendritic Cells/pathology , Histiocytosis, Langerhans-Cell/pathology , Mutation , Myeloid Progenitor Cells/pathology , Proto-Oncogene Proteins B-raf/genetics , Bone Marrow/metabolism , Dendritic Cells/metabolism , Histiocytosis, Langerhans-Cell/genetics , Histiocytosis, Langerhans-Cell/metabolism , Humans , Myeloid Progenitor Cells/metabolism
16.
J BUON ; 25(2): 612-617, 2020.
Article in English | MEDLINE | ID: mdl-32521842

ABSTRACT

Langerhans cell histiocytosis (LCH) is disease process characterized by clonal proliferation of CD1a+ dendritic cells within an inflammatory infiltrate of hematopoietic derived cells. LCH can manifest with a broad spectrum of symptoms and can involve single organs or have a multisystem distribution. Central nervous system (CNS) involvement of LCH can manifest as granulomatous parenchymal or pituitary mass lesions. Focal, space-occupying lesions, such as masses in the meninges, choroid plexus, and brain parenchyma may contain CD1a+ LCH cells, lymphocytes, and macrophages with histology similar to that of extracranial lesions. Here, we describe a rare case of multisystem LCH in an adult patient presenting with spinal lesions and isolated adrenocorticotropic (ACTH) deficiency without diabetes insipidus (DI). In addition, we review the literature summarizing the few reports of hypopituitarism in LCH in the absence of DI.


Subject(s)
Adrenocorticotropic Hormone/deficiency , Histiocytosis, Langerhans-Cell/metabolism , Histiocytosis, Langerhans-Cell/pathology , Lung/pathology , Pituitary Gland/pathology , Spine/pathology , Humans , Male , Middle Aged
17.
Ann Diagn Pathol ; 47: 151536, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32454442

ABSTRACT

Langerhans cell histiocytosis (LCH) is a bone marrow-derived immature myeloid dendritic cell proliferative disorder with diverse clinical manifestations commonly involves bone, skin, lymph node and lung. Oral involvement is uncommon. Intraoral lesions can be the first sign of either a localized LCH or clinically undiagnosed systemic LCH, predates systemic manifestations of LCH, or an early indicator of recurrence in known cases. Clinically, it can be mistaken for primary oral and dental inflammatory, infectious and neoplastic lesions. Histologically, diagnostic challenges may arise because of the nature of oral and dental specimens, different tissue reaction patterns and variations in histomorphology of LCH. We performed a retrospective review study over 10 years. We searched for diagnosed cases of LCH. We retrieved and reviewed cases of LCH with oral involvement. We found 54 cases of LCH, four (7.4%) with oral involvement. The age range was between 1 and 27 years with an average age of 13.7 years. They were males. They were clinically confused with abscess, cysts, infection, granulation tissue and other neoplastic lesions. Histologically, they showed different histopathologic features including different patterns of necrosis, granulomas, allergic-like inflammation, superimposed infection, stomatitis, cyst and sinus formation, foreign body giant cell reaction, and foci mimicking lymphomas and metastasis. Certain cytologic features were helpful hints. In doubtful cases, immunohistochemistry helped confirm the diagnosis. Because of the multiple fragmented nature of oral specimens with different tissue reaction patterns, the diagnostic Langerhans cells may be missed or misinterpreted. Oral LCH may be confused with infectious, inflammatory, benign and malignant neoplastic lesions because of its variable clinical presentations and its heterogeneous histomorphologic features. Pathologists have an important role in guiding clinicians to the correct diagnosis and patients' management. They should be familiar with the different histomorphologic patterns to avoid pitfalls. Attention to certain morphologic features and immunohistochemistry should help resolve challenging cases.


Subject(s)
Bone Marrow/pathology , Histiocytosis, Langerhans-Cell/pathology , Mouth Diseases/pathology , Adolescent , Adult , Child , Child, Preschool , Diagnosis, Differential , Giant Cells/pathology , Gingiva/pathology , Granuloma/pathology , Histiocytosis, Langerhans-Cell/diagnosis , Histiocytosis, Langerhans-Cell/metabolism , Humans , Immunohistochemistry/methods , Infant , Male , Necrosis/pathology , Pathologists/ethics , Retrospective Studies , Stomatognathic Diseases/pathology , Young Adult
18.
Cancer Immunol Res ; 8(6): 829-841, 2020 06.
Article in English | MEDLINE | ID: mdl-32238382

ABSTRACT

Langerhans cell histiocytosis (LCH) is a rare disorder characterized by tissue accumulation of CD1a+CD207+ LCH cells. In LCH, somatic mutations of the BRAF V600E gene have been detected in tissue LCH cells, bone marrow CD34+ hematopoietic stem cells, circulating CD14+ monocytes, and BDCA1+ myeloid dendritic cells (DC). Targeting BRAF V600E in clonal Langerhans cells (LC) and their precursors is a potential treatment option for patients whose tumors have the mutation. The development of mouse macrophages and LCs is regulated by the CSF1 receptor (CSF1R). In patients with diffuse-type tenosynovial giant cell tumors, CSF1R inhibition depletes tumor-associated macrophages (TAM) with therapeutic efficacy; however, CSF1R signaling in LCs and LCH has not been investigated. We found through IHC and flow cytometry that CSF1R is normally expressed on human CD1a+CD207+ LCs in the epidermis and stratified epithelia. LCs that were differentiated from CD14+ monocytes, BDCA1+ DCs, and CD34+ cord blood progenitors expressed CSF1R that was downregulated upon maturation. Immature LCs migrated toward CSF1, but not IL34. Administration of the c-FMS/CSF1R kinase inhibitors GW2580 and BLZ945 significantly reduced human LC migration. In LCH clinical samples, LCH cells (including BRAF V600E cells) and TAMs retained high expression of CSF1R. We also detected the presence of transcripts for its ligand, CSF1, but not IL34, in all tested LCH cases. CSF1R and CSF1 expression in LCH, and their role in LC migration and differentiation, suggests CSF1R signaling blockade as a candidate rational approach for treatment of LCH, including the BRAF V600E and wild-type forms of the disease.


Subject(s)
Cell Differentiation , Cell Movement , Dendritic Cells/pathology , Histiocytosis, Langerhans-Cell/pathology , Langerhans Cells/pathology , Monocytes/pathology , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Adolescent , Adult , Aged , Apoptosis , Cell Lineage , Cells, Cultured , Child , Child, Preschool , Dendritic Cells/metabolism , Female , Histiocytosis, Langerhans-Cell/metabolism , Humans , Infant , Infant, Newborn , Langerhans Cells/metabolism , Male , Middle Aged , Monocytes/metabolism , Tumor Microenvironment , Young Adult
19.
Cancer Discov ; 9(10): 1406-1421, 2019 10.
Article in English | MEDLINE | ID: mdl-31345789

ABSTRACT

Langerhans cell histiocytosis (LCH) is a rare neoplasm predominantly affecting children. It occupies a hybrid position between cancers and inflammatory diseases, which makes it an attractive model for studying cancer development. To explore the molecular mechanisms underlying the pathophysiology of LCH and its characteristic clinical heterogeneity, we investigated the transcriptomic and epigenomic diversity in primary LCH lesions. Using single-cell RNA sequencing, we identified multiple recurrent types of LCH cells within these biopsies, including putative LCH progenitor cells and several subsets of differentiated LCH cells. We confirmed the presence of proliferative LCH cells in all analyzed biopsies using IHC, and we defined an epigenomic and gene-regulatory basis of the different LCH-cell subsets by chromatin-accessibility profiling. In summary, our single-cell analysis of LCH uncovered an unexpected degree of cellular, transcriptomic, and epigenomic heterogeneity among LCH cells, indicative of complex developmental hierarchies in LCH lesions. SIGNIFICANCE: This study sketches a molecular portrait of LCH lesions by combining single-cell transcriptomics with epigenome profiling. We uncovered extensive cellular heterogeneity, explained in part by an intrinsic developmental hierarchy of LCH cells. Our findings provide new insights and hypotheses for advancing LCH research and a starting point for personalizing therapy.See related commentary by Gruber et al., p. 1343.This article is highlighted in the In This Issue feature, p. 1325.


Subject(s)
Epigenesis, Genetic , Epigenomics , Histiocytosis, Langerhans-Cell/genetics , Biomarkers , Biopsy , Disease Susceptibility , Epigenomics/methods , Gene Expression Profiling , Gene Expression Regulation , High-Throughput Nucleotide Sequencing , Histiocytosis, Langerhans-Cell/diagnosis , Histiocytosis, Langerhans-Cell/metabolism , Humans , Immunohistochemistry , Radiography , Single-Cell Analysis
20.
BMC Cancer ; 19(1): 514, 2019 May 29.
Article in English | MEDLINE | ID: mdl-31142285

ABSTRACT

BACKGROUND: Crystal-storing histiocytosis (CSH) is a rare lesion characterized by sheets of crystal-laden non-neoplastic histiocytes. CSH shows a prominent association with lymphoproliferative disorders that express monoclonal immunoglobulins, mainly multiple myeloma (MM), lymphoplasmacytic lymphoma (LPL) and monoclonal gammopathy of undetermined significance (MGUS). However, no aggressive B cell lymphoma has been reported to be associated with CSH. CASE PRESENTATION: A 74-year-old Chinese woman presented with multiple subcutaneous masses, abdominal pain, and fever. An IgM kappa type of monoclonal gammopathy (MG) was noted by immunofixation performed on the patient's serum. Computed tomographic (CT) scan revealed subcutaneous masses on the left upper arm and at the waist and multiple low-density lesions in the spleen. Microscopically, sections of subcutaneous masses revealed sheets of large polygonal and spindle cells with abundant eosinophilic cytoplasm, round to ovoid eccentric nuclei, reticulate chromatin, and median nucleoli. Massive needle-shaped crystals were confined to the cytoplasm. Immunohistochemically, these crystal-containing cells were positive for CD68/PGM1, CD163, IgM, and Igκ. Meanwhile, the splenic tumour was diagnosed as diffuse large B-cell lymphoma (DLBCL), non-germinal-centre B (non-GCB) subtype (Hans algorithm). Immunohistochemistry for IgM was positive in the cytoplasm of some neoplastic cells. Immunoglobulin heavy chain (IgH) gene rearrangement was detected by PCR analysis of the subcutaneous mass and the splenic tumour. CONCLUSION: To the best of our knowledge, this is the first case of generalized CSH with DLBCL and MG. Although the rarity of CSH and separate locations of CSH and lymphoma led to a diagnostic dilemma, the presence of MG was a clue to appreciate the relation between CSH and DLBCL. This case stressed a full investigation into the underlying lymphoproliferative disorder for integrated diagnosis and correct treatments.


Subject(s)
Histiocytosis, Langerhans-Cell/diagnostic imaging , Lymphoma, Large B-Cell, Diffuse/diagnostic imaging , Paraproteinemias/diagnostic imaging , Aged , Female , Gene Rearrangement , Histiocytosis, Langerhans-Cell/genetics , Histiocytosis, Langerhans-Cell/metabolism , Humans , Immunoglobulin Heavy Chains/genetics , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/metabolism , Paraproteinemias/genetics , Paraproteinemias/metabolism , Tomography, X-Ray Computed
SELECTION OF CITATIONS
SEARCH DETAIL
...