Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 287
Filtrar
1.
Clin Immunol ; 263: 110203, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38575046

RESUMO

Langerhans cell histiocytosis (LCH) is characterized by an expansion and accumulation of pathological histiocytes expressing langerin (CD207) and CD1a in different organs under an inflammatory milieu. The origin of pathognomonic precursors of LCH is widely debated, but monocytes and pre-dendritic cells (pre-DC) play a significant role. Remarkably, we found an expansion of AXLhigh cells in the CD11c+ subset of patients with active LCH, which also express the pathognomonic CD207 and CD1a. Moreover, we obtained a monocyte-derived LC-like (mo-LC-like) expressing high levels of AXL when treated with inflammatory cytokine, or plasma of patients with active disease. Intriguingly, inhibiting the mTOR pathway at the initial stages of monocyte differentiation to LC-like fosters the pathognomonic LCH program, highly increasing CD207 levels, together with NOTCH1 induction. We define here that AXLhigh could also be taken as a strong pathognomonic marker for LCH, and the release of Langerin and NOTCH1 expression depends on the inhibition of the mTOR pathway.


Assuntos
Antígenos CD , Receptor Tirosina Quinase Axl , Histiocitose de Células de Langerhans , Lectinas Tipo C , Lectinas de Ligação a Manose , Proteínas Proto-Oncogênicas , Receptores Proteína Tirosina Quinases , Serina-Treonina Quinases TOR , Humanos , Histiocitose de Células de Langerhans/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Antígenos CD/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas/genética , Lectinas de Ligação a Manose/metabolismo , Lectinas Tipo C/metabolismo , Masculino , Células Mieloides/metabolismo , Biomarcadores , Feminino , Adolescente , Receptor Notch1/metabolismo , Antígenos CD1/metabolismo , Criança , Monócitos/metabolismo , Monócitos/imunologia , Adulto , Pré-Escolar , Transdução de Sinais , Diferenciação Celular
2.
Immunity ; 56(12): 2677-2678, 2023 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-38091948

RESUMO

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.


Assuntos
Histiocitose de Células de Langerhans , Monócitos , Humanos , Monócitos/metabolismo , Histiocitose de Células de Langerhans/metabolismo , Encéfalo/metabolismo , Barreira Hematoencefálica/metabolismo , Células Mieloides/metabolismo , Proteínas Proto-Oncogênicas B-raf/metabolismo
3.
Cancer Sci ; 114(5): 1871-1881, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36788737

RESUMO

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.


Assuntos
Histiocitose de Células de Langerhans , Animais , Humanos , Camundongos , Histiocitose de Células de Langerhans/tratamento farmacológico , Histiocitose de Células de Langerhans/genética , Histiocitose de Células de Langerhans/metabolismo , Baço/metabolismo
4.
Sci Immunol ; 7(78): eadd3330, 2022 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-36525505

RESUMO

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.


Assuntos
Histiocitose de Células de Langerhans , Neoplasias , Humanos , Linhagem da Célula , Histiocitose de Células de Langerhans/metabolismo , Histiocitose de Células de Langerhans/patologia , Diferenciação Celular , Monócitos/metabolismo
5.
Vnitr Lek ; 68(E-2): 11-21, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36208940

RESUMO

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.


Assuntos
Histiocitose de Células de Langerhans , Adulto , Histiocitose de Células de Langerhans/diagnóstico , Histiocitose de Células de Langerhans/metabolismo , Histiocitose de Células de Langerhans/terapia , Humanos , Linfonodos/patologia , Doenças Raras
6.
J Leukoc Biol ; 112(5): 1089-1104, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35648675

RESUMO

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.


Assuntos
Histiocitose de Células de Langerhans , Proteínas Proto-Oncogênicas B-raf , Camundongos , Animais , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas B-raf/metabolismo , Lipopolissacarídeos/farmacologia , Fator de Necrose Tumoral alfa/genética , Receptor 4 Toll-Like/genética , Mutação , Histiocitose de Células de Langerhans/metabolismo , Células Dendríticas/metabolismo
7.
J Immunol ; 209(2): 270-279, 2022 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-35768149

RESUMO

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.


Assuntos
Histiocitose de Células de Langerhans , Lectinas de Ligação a Manose , Antígenos CD/metabolismo , Antígenos CD1/metabolismo , Histiocitose de Células de Langerhans/diagnóstico , Histiocitose de Células de Langerhans/metabolismo , Humanos , Células de Langerhans , Lectinas Tipo C/metabolismo , Lectinas de Ligação a Manose/metabolismo
8.
Am J Ophthalmol ; 242: 36-51, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35594918

RESUMO

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.


Assuntos
Histiocitose de Células de Langerhans , Neoplasias , Adulto , Ciclina D1/genética , Células Dendríticas/metabolismo , Células Dendríticas/patologia , Feminino , Histiocitose de Células de Langerhans/genética , Histiocitose de Células de Langerhans/metabolismo , Histiocitose de Células de Langerhans/patologia , Humanos , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Proteínas Quinases Ativadas por Mitógeno , Biologia Molecular , Estudos Retrospectivos
9.
Tohoku J Exp Med ; 255(1): 27-31, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34497165

RESUMO

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.


Assuntos
Histiocitose de Células de Langerhans/metabolismo , Sarcoma de Células de Langerhans/metabolismo , Proteínas de Ligação a RNA/metabolismo , Adolescente , Adulto , Biomarcadores/metabolismo , Biomarcadores Tumorais/metabolismo , Criança , Pré-Escolar , Diagnóstico Diferencial , Feminino , Histiocitose de Células de Langerhans/diagnóstico , Humanos , Imuno-Histoquímica , Lactente , Recém-Nascido , Sarcoma de Células de Langerhans/diagnóstico , Masculino , Pessoa de Meia-Idade , Adulto Jovem
10.
Ann Diagn Pathol ; 52: 151720, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33676171

RESUMO

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.


Assuntos
Histiocitose de Células de Langerhans/diagnóstico , Sarcoma de Células de Langerhans/diagnóstico , Linfoma Folicular/diagnóstico , Segunda Neoplasia Primária/patologia , Adulto , Idoso , Linfócitos B/patologia , Desdiferenciação Celular/genética , Transdiferenciação Celular/genética , Feminino , Histiócitos/patologia , Histiocitose de Células de Langerhans/metabolismo , Histiocitose de Células de Langerhans/patologia , Humanos , Imuno-Histoquímica/métodos , Imunofenotipagem/métodos , Sarcoma de Células de Langerhans/metabolismo , Sarcoma de Células de Langerhans/patologia , Células de Langerhans/patologia , Linfoma Folicular/metabolismo , Linfoma Folicular/patologia , Masculino , Pessoa de Meia-Idade , Proteínas Quinases Ativadas por Mitógeno/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Estadiamento de Neoplasias/métodos
11.
Pathol Int ; 71(3): 191-198, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33497038

RESUMO

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.


Assuntos
Histiocitose de Células de Langerhans , Antígenos de Histocompatibilidade Classe I/metabolismo , Imunoglobulinas Intravenosas , Receptores Fc/metabolismo , Adolescente , Adulto , Albuminas/metabolismo , Linhagem Celular , Criança , Pré-Escolar , Feminino , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/metabolismo , Histiocitose de Células de Langerhans/tratamento farmacológico , Histiocitose de Células de Langerhans/metabolismo , Humanos , Imunoglobulinas Intravenosas/metabolismo , Imunoglobulinas Intravenosas/farmacologia , Lactente , Recém-Nascido , Masculino , Albumina Sérica/metabolismo
12.
Am J Surg Pathol ; 44(12): 1658-1665, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32910018

RESUMO

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.


Assuntos
Biomarcadores Tumorais/genética , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/patologia , Histiocitose de Células de Langerhans/genética , Histiocitose de Células de Langerhans/patologia , Neoplasias Renais/genética , Neoplasias Renais/patologia , Mutação , Proteínas Proto-Oncogênicas B-raf/genética , Adulto , Idoso , Biomarcadores Tumorais/análise , Carcinoma de Células Renais/química , Carcinoma de Células Renais/cirurgia , Feminino , Histiocitose de Células de Langerhans/metabolismo , Histiocitose de Células de Langerhans/cirurgia , Humanos , Neoplasias Renais/química , Neoplasias Renais/cirurgia , Masculino , Pessoa de Meia-Idade , Gradação de Tumores , Nefrectomia , Estudos Prospectivos
13.
Am J Med Genet A ; 182(11): 2746-2750, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32945094

RESUMO

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.


Assuntos
Histiocitose de Células de Langerhans/patologia , Hiperventilação/complicações , Deficiência Intelectual/complicações , Mutação , Fator de Transcrição 4/genética , Criança , Fácies , Histiocitose de Células de Langerhans/etiologia , Histiocitose de Células de Langerhans/metabolismo , Humanos , Masculino , Fenótipo
15.
Scand J Immunol ; 92(6): e12950, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32738155

RESUMO

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.


Assuntos
Histiocitose de Células de Langerhans/metabolismo , Células de Langerhans/fisiologia , Macrófagos/metabolismo , Linfócitos T Reguladores/metabolismo , Adulto , Antígenos CD/metabolismo , Diferenciação Celular , Microambiente Celular , Criança , Pré-Escolar , Citocinas/metabolismo , Células Dendríticas/metabolismo , Feminino , Fatores de Transcrição Forkhead/metabolismo , Humanos , Imuno-Histoquímica/métodos , Lactente , Macrófagos/imunologia , Masculino , Linfócitos T Citotóxicos/metabolismo , Linfócitos T Reguladores/imunologia , Células Th2/imunologia
16.
Blood ; 136(19): 2188-2199, 2020 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-32750121

RESUMO

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.


Assuntos
Medula Óssea/patologia , Diferenciação Celular , Células Dendríticas/patologia , Histiocitose de Células de Langerhans/patologia , Mutação , Células Progenitoras Mieloides/patologia , Proteínas Proto-Oncogênicas B-raf/genética , Medula Óssea/metabolismo , Células Dendríticas/metabolismo , Histiocitose de Células de Langerhans/genética , Histiocitose de Células de Langerhans/metabolismo , Humanos , Células Progenitoras Mieloides/metabolismo
17.
J BUON ; 25(2): 612-617, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32521842

RESUMO

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.


Assuntos
Hormônio Adrenocorticotrópico/deficiência , Histiocitose de Células de Langerhans/metabolismo , Histiocitose de Células de Langerhans/patologia , Pulmão/patologia , Hipófise/patologia , Coluna Vertebral/patologia , Humanos , Masculino , Pessoa de Meia-Idade
18.
Ann Diagn Pathol ; 47: 151536, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32454442

RESUMO

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.


Assuntos
Medula Óssea/patologia , Histiocitose de Células de Langerhans/patologia , Doenças da Boca/patologia , Adolescente , Adulto , Criança , Pré-Escolar , Diagnóstico Diferencial , Células Gigantes/patologia , Gengiva/patologia , Granuloma/patologia , Histiocitose de Células de Langerhans/diagnóstico , Histiocitose de Células de Langerhans/metabolismo , Humanos , Imuno-Histoquímica/métodos , Lactente , Masculino , Necrose/patologia , Patologistas/ética , Estudos Retrospectivos , Doenças Estomatognáticas/patologia , Adulto Jovem
19.
Cancer Immunol Res ; 8(6): 829-841, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32238382

RESUMO

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.


Assuntos
Diferenciação Celular , Movimento Celular , Células Dendríticas/patologia , Histiocitose de Células de Langerhans/patologia , Células de Langerhans/patologia , Monócitos/patologia , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Adolescente , Adulto , Idoso , Apoptose , Linhagem da Célula , Células Cultivadas , Criança , Pré-Escolar , Células Dendríticas/metabolismo , Feminino , Histiocitose de Células de Langerhans/metabolismo , Humanos , Lactente , Recém-Nascido , Células de Langerhans/metabolismo , Masculino , Pessoa de Meia-Idade , Monócitos/metabolismo , Microambiente Tumoral , Adulto Jovem
20.
Cancer Discov ; 9(10): 1406-1421, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31345789

RESUMO

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.


Assuntos
Epigênese Genética , Epigenômica , Histiocitose de Células de Langerhans/genética , Biomarcadores , Biópsia , Suscetibilidade a Doenças , Epigenômica/métodos , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Histiocitose de Células de Langerhans/diagnóstico , Histiocitose de Células de Langerhans/metabolismo , Humanos , Imuno-Histoquímica , Radiografia , Análise de Célula Única
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...