Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Cell ; 186(21): 4546-4566.e27, 2023 Oct 12.
Article in English | MEDLINE | ID: mdl-37769657

ABSTRACT

Neutrophils are abundant immune cells in the circulation and frequently infiltrate tumors in substantial numbers. However, their precise functions in different cancer types remain incompletely understood, including in the brain microenvironment. We therefore investigated neutrophils in tumor tissue of glioma and brain metastasis patients, with matched peripheral blood, and herein describe the first in-depth analysis of neutrophil phenotypes and functions in these tissues. Orthogonal profiling strategies in humans and mice revealed that brain tumor-associated neutrophils (TANs) differ significantly from blood neutrophils and have a prolonged lifespan and immune-suppressive and pro-angiogenic capacity. TANs exhibit a distinct inflammatory signature, driven by a combination of soluble inflammatory mediators including tumor necrosis factor alpha (TNF-ɑ) and Ceruloplasmin, which is more pronounced in TANs from brain metastasis versus glioma. Myeloid cells, including tumor-associated macrophages, emerge at the core of this network of pro-inflammatory mediators, supporting the concept of a critical myeloid niche regulating overall immune suppression in human brain tumors.

2.
Cell Rep Med ; 4(1): 100900, 2023 01 17.
Article in English | MEDLINE | ID: mdl-36652909

ABSTRACT

Brain metastases (BrMs) are the most common form of brain tumors in adults and frequently originate from lung and breast primary cancers. BrMs are associated with high mortality, emphasizing the need for more effective therapies. Genetic profiling of primary tumors is increasingly used as part of the effort to guide targeted therapies against BrMs, and immune-based strategies for the treatment of metastatic cancer are gaining momentum. However, the tumor immune microenvironment (TIME) of BrM is extremely heterogeneous, and whether specific genetic profiles are associated with distinct immune states remains unknown. Here, we perform an extensive characterization of the immunogenomic landscape of human BrMs by combining whole-exome/whole-genome sequencing, RNA sequencing of immune cell populations, flow cytometry, immunofluorescence staining, and tissue imaging analyses. This revealed unique TIME phenotypes in genetically distinct lung- and breast-BrMs, thereby enabling the development of personalized immunotherapies tailored by the genetic makeup of the tumors.


Subject(s)
Brain Neoplasms , Breast Neoplasms , Melanoma , Skin Neoplasms , Adult , Humans , Female , Brain Neoplasms/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Immunotherapy , Tumor Microenvironment/genetics
3.
Inflamm Bowel Dis ; 29(10): 1586-1593, 2023 10 03.
Article in English | MEDLINE | ID: mdl-36617284

ABSTRACT

BACKGROUND: Carriers of the human leucocyte antigen variant HLADQA1*05 (rs2097432) are at risk of developing antibodies against infliximab and adalimumab with reduced tumor necrosis factor (TNF) antagonist persistence. The impact of proactive therapeutic drug monitoring (PTDM) on this association has been barely assessed. METHODS: We conducted a retrospective single-center cohort study including patients with inflammatory bowel disease starting anti-TNF therapy between January 2017 and March 2021. Proactive therapeutic drug monitoring was defined as periodic drug level measurement (≥2 determinations during the first year of treatment and ≥1/annual determination during the following years), regardless of clinical condition, followed by dose optimization. Variables associated with treatment persistence were assessed with multivariable Cox regression analysis. RESULTS: A total of 112 patients were included, 52 (46.4%) HLA-DQA1*05 carriers, with a median follow-up of 73.9 (interquartile range, 35.4-133.1) weeks. Combination therapy with thiopurines was more frequent among HLA-DQA1*05 noncarriers (28 [46.7%] vs 12 [23.1%]; P = .01). Clinical remission rates at week 14 (77.9% vs 73.9%; P = .69) and 56 (73.2% vs 68.4%; P = .64) were similar between HLA-DQA1*05 noncarriers and carriers. Drug persistence was higher among HLA-DQA1*05 carriers (hazard ratio [HR], 0.32; 95% confidence interval, 0.14-0.71; P = .01). Multivariable Cox regression analysis identified systemic steroids at anti-TNF initiation (HR, 4; 95% confidence interval, 1.7-9.7) as a risk factor and HLA-DQA1*05 carriers (HR, 0.31; 95% confidence interval, 0.12-0.81) as a protective factor of treatment cessation. CONCLUSION: In adult patients with PTDM, a positive HLA-DQA1*05 genotype does not associate a higher risk of treatment cessation nor worse clinical outcomes.


This is a retrospective cohort study including 112 inflammatory bowel disease patients starting anti-TNF therapy under proactive therapeutic drug monitoring (PTDM). The HLA-DQA1*05 carriers did not present lower drug persistence or remission rates, suggesting PTDM overcomes the reduced treatment survival expected in HLA-DQA1*05 carriers.


Subject(s)
Inflammatory Bowel Diseases , Tumor Necrosis Factor Inhibitors , Humans , Adult , Tumor Necrosis Factor Inhibitors/therapeutic use , Retrospective Studies , Cohort Studies , Drug Monitoring , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/genetics , Infliximab , Adalimumab/therapeutic use , Genotype , Tumor Necrosis Factor-alpha
4.
Nat Protoc ; 16(10): 4692-4721, 2021 10.
Article in English | MEDLINE | ID: mdl-34462595

ABSTRACT

Human tissue samples represent an invaluable source of information for the analysis of disease-specific cellular alterations and their variation between different pathologies. In cancer research, advancing a comprehensive understanding of the unique characteristics of individual tumor types and their microenvironment is of considerable importance for clinical translation. However, investigating human brain tumor tissue is challenging due to the often-limited availability of surgical specimens. Here we describe a multimodule integrated pipeline for the processing of freshly resected human brain tumor tissue and matched blood that enables analysis of the tumor microenvironment, with a particular focus on the tumor immune microenvironment (TIME). The protocol maximizes the information yield from limited tissue and includes both the preservation of bulk tissue, which can be performed within 1 h following surgical resection, as well as tissue dissociation for an in-depth characterization of individual TIME cell populations, which typically takes several hours depending on tissue quantity and further downstream processing. We also describe integrated modules for immunofluorescent staining of sectioned tissue, bulk tissue genomic analysis and fluorescence- or magnetic-activated cell sorting of digested tissue for subsequent culture or transcriptomic analysis by RNA sequencing. Applying this pipeline, we have previously described the overall TIME landscape across different human brain malignancies, and were able to delineate disease-specific alterations of tissue-resident versus recruited macrophage populations. This protocol will enable researchers to use this pipeline to address further research questions regarding the tumor microenvironment.


Subject(s)
Brain Neoplasms , Gene Expression Profiling , Humans , Macrophages , Sequence Analysis, RNA , Tumor Microenvironment
5.
Nucleic Acids Res ; 49(9): 5057-5073, 2021 05 21.
Article in English | MEDLINE | ID: mdl-33950194

ABSTRACT

Activation-induced deaminase (AID) initiates antibody diversification in germinal center B cells by deaminating cytosines, leading to somatic hypermutation and class-switch recombination. Loss-of-function mutations in AID lead to hyper-IgM syndrome type 2 (HIGM2), a rare human primary antibody deficiency. AID-mediated deamination has been proposed as leading to active demethylation of 5-methycytosines in the DNA, although evidence both supports and casts doubt on such a role. In this study, using whole-genome bisulfite sequencing of HIGM2 B cells, we investigated direct AID involvement in active DNA demethylation. HIGM2 naïve and memory B cells both display widespread DNA methylation alterations, of which ∼25% are attributable to active DNA demethylation. For genes that undergo active demethylation that is impaired in HIGM2 individuals, our analysis indicates that AID is not directly involved. We demonstrate that the widespread alterations in the DNA methylation and expression profiles of HIGM2 naïve B cells result from premature overstimulation of the B-cell receptor prior to the germinal center reaction. Our data support a role for AID in B cell central tolerance in preventing the expansion of autoreactive cell clones, affecting the correct establishment of DNA methylation patterns.


Subject(s)
B-Lymphocytes/immunology , Cytidine Deaminase/physiology , DNA Methylation , Hyper-IgM Immunodeficiency Syndrome/genetics , Hyper-IgM Immunodeficiency Syndrome/immunology , Autoimmunity , B-Lymphocytes/metabolism , Cytidine Deaminase/deficiency , Cytidine Deaminase/genetics , Germinal Center/immunology , Humans , Hyper-IgM Immunodeficiency Syndrome/metabolism , Immune Tolerance , Immunologic Memory , Receptors, Antigen, B-Cell/genetics , Transcriptome , Whole Genome Sequencing
6.
Int Rev Cell Mol Biol ; 358: 85-104, 2021.
Article in English | MEDLINE | ID: mdl-33707058

ABSTRACT

Dendritic cells (DC) and macrophages (Mϕ) constitute the most abundant antigen presenting cells in the human intestinal mucosa. In resting conditions, they are essential to maintain the mechanisms of immune tolerance toward food antigens and commensals, at the time that they keep the capacity to initiate and maintain antigen-specific pro-inflammatory immune responses toward invading pathogens. Nevertheless, this delicate equilibrium between immunity and tolerance is not perfect, like in coeliac disease (CD), where DC and Mϕ drive the development of antigen-specific immune responses toward dietary gluten peptides. In this review, we provide therefore a comprehensive discussion about CD pathogenesis, the human intestinal immune system and the biology of intestinal DC and Mϕ both in resting conditions and in CD. Last, but not least, we discuss about all the remaining issues pending to be studied regarding DC and Mϕ contribution toward CD pathogenesis. This may allow the identification of unique and specific factors which may be useful in the clinical practice, as well as identify new therapeutic targets in order to reestablish the loss intestinal homeostasis in CD.


Subject(s)
Celiac Disease/pathology , Dendritic Cells/pathology , Intestines/pathology , Macrophages/pathology , Animals , Humans , Immune Tolerance , Immunity, Innate , Intestines/immunology
7.
PLoS Genet ; 16(12): e1008960, 2020 12.
Article in English | MEDLINE | ID: mdl-33362210

ABSTRACT

Most B cell lymphomas originate from B cells that have germinal center (GC) experience and bear chromosome translocations and numerous point mutations. GC B cells remodel their immunoglobulin (Ig) genes by somatic hypermutation (SHM) and class switch recombination (CSR) in their Ig genes. Activation Induced Deaminase (AID) initiates CSR and SHM by generating U:G mismatches on Ig DNA that can then be processed by Uracyl-N-glycosylase (UNG). AID promotes collateral damage in the form of chromosome translocations and off-target SHM, however, the exact contribution of AID activity to lymphoma generation and progression is not completely understood. Here we show using a conditional knock-in strategy that AID supra-activity alone is not sufficient to generate B cell transformation. In contrast, in the absence of UNG, AID supra-expression increases SHM and promotes lymphoma. Whole exome sequencing revealed that AID heavily contributes to lymphoma SHM, promoting subclonal variability and a wider range of oncogenic variants. Thus, our data provide direct evidence that UNG is a brake to AID-induced intratumoral heterogeneity and evolution of B cell lymphoma.


Subject(s)
Cytidine Deaminase/genetics , Genetic Heterogeneity , Lymphoma, B-Cell/genetics , Uracil-DNA Glycosidase/genetics , Animals , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Clonal Evolution , Cytidine Deaminase/metabolism , Female , Lymphoma, B-Cell/metabolism , Male , Mice , Mice, Inbred C57BL , Mutation , Uracil-DNA Glycosidase/metabolism
8.
Leukemia ; 34(10): 2722-2735, 2020 10.
Article in English | MEDLINE | ID: mdl-32576963

ABSTRACT

Mutations in genes encoding subunits of the SWI/SNF chromatin remodeling complex are frequently found in different human cancers. While the tumor suppressor function of this complex is widely established in solid tumors, its role in hematologic malignancies is largely unknown. Recurrent point mutations in BCL7A gene, encoding a subunit of the SWI/SNF complex, have been reported in diffuse large B-cell lymphoma (DLBCL), but their functional impact remains to be elucidated. Here we show that BCL7A often undergoes biallelic inactivation, including a previously unnoticed mutational hotspot in the splice donor site of intron one. The splice site mutations render a truncated BCL7A protein, lacking a portion of the amino-terminal domain. Moreover, restoration of wild-type BCL7A expression elicits a tumor suppressor-like phenotype in vitro and in vivo. In contrast, splice site mutations block the tumor suppressor function of BCL7A by preventing its binding to the SWI/SNF complex. We also show that BCL7A restoration induces transcriptomic changes in genes involved in B-cell activation. In addition, we report that SWI/SNF complex subunits harbor mutations in more than half of patients with germinal center B-cell (GCB)-DLBCL. Overall, this work demonstrates the tumor suppressor function of BCL7A in DLBCL, and highlights that the SWI/SNF complex plays a relevant role in DLBCL pathogenesis.


Subject(s)
Genes, Tumor Suppressor , Lymphoma, Large B-Cell, Diffuse/genetics , Microfilament Proteins/genetics , Mutation , Oncogene Proteins/genetics , Protein Interaction Domains and Motifs/genetics , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Chromatography, Liquid , Chromosomal Proteins, Non-Histone/metabolism , DNA Mutational Analysis , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Lymphocyte Activation/immunology , Lymphoma, Large B-Cell, Diffuse/diagnosis , Lymphoma, Large B-Cell, Diffuse/therapy , Mice , Microfilament Proteins/chemistry , Molecular Imaging , Multiprotein Complexes , Oncogene Proteins/chemistry , Protein Binding , Tandem Mass Spectrometry , Xenograft Model Antitumor Assays
9.
Nat Commun ; 10(1): 5563, 2019 12 05.
Article in English | MEDLINE | ID: mdl-31804490

ABSTRACT

The prerequisite to prevent childhood B-cell acute lymphoblastic leukemia (B-ALL) is to decipher its etiology. The current model suggests that infection triggers B-ALL development through induction of activation-induced cytidine deaminase (AID; also known as AICDA) in precursor B-cells. This evidence has been largely acquired through the use of ex vivo functional studies. However, whether this mechanism governs native non-transplant B-ALL development is unknown. Here we show that, surprisingly, AID genetic deletion does not affect B-ALL development in Pax5-haploinsufficient mice prone to B-ALL upon natural infection exposure. We next test the effect of premature AID expression from earliest pro-B-cell stages in B-cell transformation. The generation of AID off-target mutagenic activity in precursor B-cells does not promote B-ALL. Likewise, known drivers of human B-ALL are not preferentially targeted by AID. Overall these results suggest that infections promote B-ALL through AID-independent mechanisms, providing evidence for a new model of childhood B-ALL development.


Subject(s)
B-Lymphocytes/metabolism , Cell Transformation, Neoplastic/metabolism , Cytidine Deaminase/metabolism , Infections/physiopathology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Animals , B-Lymphocytes/pathology , Cell Transformation, Neoplastic/genetics , Child , Cytidine Deaminase/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , High-Throughput Nucleotide Sequencing/methods , Humans , Infections/genetics , Kaplan-Meier Estimate , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , PAX5 Transcription Factor/genetics , PAX5 Transcription Factor/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics
10.
J Exp Med ; 215(3): 761-771, 2018 03 05.
Article in English | MEDLINE | ID: mdl-29374026

ABSTRACT

Activation-induced deaminase (AID) initiates antibody diversification in germinal center (GC) B cells through the deamination of cytosines on immunoglobulin genes. AID can also target other regions in the genome, triggering mutations or chromosome translocations, with major implications for oncogenic transformation. However, understanding the specificity of AID has proved extremely challenging. We have sequenced at very high depth >1,500 genomic regions from GC B cells and identified 275 genes targeted by AID, including 30 of the previously known 35 AID targets. We have also identified the most highly mutated hotspot for AID activity described to date. Furthermore, integrative analysis of the molecular features of mutated genes coupled to machine learning has produced a powerful predictive tool for AID targets. We also have found that base excision repair and mismatch repair back up each other to faithfully repair AID-induced lesions. Finally, our data establish a novel link between AID mutagenic activity and lymphomagenesis.


Subject(s)
Cytidine Deaminase/genetics , Somatic Hypermutation, Immunoglobulin/genetics , Animals , Base Sequence , DNA Damage/genetics , DNA Mismatch Repair/genetics , High-Throughput Nucleotide Sequencing , Lymphoma/genetics , Lymphoma/pathology , Mice , Mutation/genetics
11.
Nat Commun ; 8: 16067, 2017 07 05.
Article in English | MEDLINE | ID: mdl-28677680

ABSTRACT

In germinal centres (GC) mature B cells undergo intense proliferation and immunoglobulin gene modification before they differentiate into memory B cells or long-lived plasma cells (PC). GC B-cell-to-PC transition involves a major transcriptional switch that promotes a halt in cell proliferation and the production of secreted immunoglobulins. Here we show that the CCCTC-binding factor (CTCF) is required for the GC reaction in vivo, whereas in vitro the requirement for CTCF is not universal and instead depends on the pathways used for B-cell activation. CTCF maintains the GC transcriptional programme, allows a high proliferation rate, and represses the expression of Blimp-1, the master regulator of PC differentiation. Restoration of Blimp-1 levels partially rescues the proliferation defect of CTCF-deficient B cells. Thus, our data reveal an essential function of CTCF in maintaining the GC transcriptional programme and preventing premature PC differentiation.


Subject(s)
B-Lymphocytes/physiology , CCCTC-Binding Factor/physiology , Cell Differentiation , Germinal Center/metabolism , Animals , Female , Male , Mice , Plasma Cells , Positive Regulatory Domain I-Binding Factor 1/metabolism , Primary Cell Culture , Transcription, Genetic
12.
Blood ; 129(17): 2408-2419, 2017 04 27.
Article in English | MEDLINE | ID: mdl-28188132

ABSTRACT

Non-Hodgkin lymphoma comprises a variety of neoplasms, many of which arise from germinal center (GC)-experienced B cells. microRNA-28 (miR-28) is a GC-specific miRNA whose expression is lost in numerous mature B-cell neoplasms. Here we show that miR-28 regulates the GC reaction in primary B cells by impairing class switch recombination and memory B and plasma cell differentiation. Deep quantitative proteomics combined with transcriptome analysis identified miR-28 targets involved in cell-cycle and B-cell receptor signaling. Accordingly, we found that miR-28 expression diminished proliferation in primary and lymphoma cells in vitro. Importantly, miR-28 reexpression in human Burkitt (BL) and diffuse large B-cell lymphoma (DLBCL) xenografts blocked tumor growth, both when delivered in viral vectors or as synthetic, clinically amenable, molecules. Further, the antitumoral effect of miR-28 is conserved in a primary murine in vivo model of BL. Thus, miR-28 replacement is uncovered as a novel therapeutic strategy for DLBCL and BL treatment.


Subject(s)
B-Lymphocytes/immunology , Burkitt Lymphoma/therapy , Gene Expression Regulation, Neoplastic , Germinal Center/immunology , Lymphoma, Large B-Cell, Diffuse/therapy , MicroRNAs/genetics , Animals , B-Lymphocytes/pathology , Burkitt Lymphoma/genetics , Burkitt Lymphoma/immunology , Burkitt Lymphoma/pathology , Cell Differentiation , Cell Proliferation , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Germinal Center/pathology , Humans , Immunoglobulin Class Switching , Immunologic Memory , Lentivirus/genetics , Lentivirus/metabolism , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/immunology , Lymphoma, Large B-Cell, Diffuse/pathology , Mice , Mice, Inbred NOD , Mice, SCID , MicroRNAs/immunology , Plasma Cells/immunology , Plasma Cells/pathology , Proteomics , Transcriptome , Transfection , Xenograft Model Antitumor Assays
14.
J Proteome Res ; 11(12): 5972-82, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-23140423

ABSTRACT

Protein interactions play a critical role in the regulation of many biological events and their study in a high-throughput format has become a key area of proteomic research. Nucleid Acid Programmable Protein Arrays (NAPPA) technology allows the construction of protein arrays from cDNA expression libraries in high-throughput cell-free systems to study protein interaction and functions. Tick saliva contains antihemostatic, anti-inflammatory, and immunosuppressive proteins that counteract the host hemostatic, immune, and inflammatory responses allowing the ingestion of host blood and facilitating its infection by the tick-borne pathogens. Identification of such proteins and their functions could help in the selection of antigenic targets for the development of antitick and transmission-blocking vaccines. With that aim, we have prepared a cDNA expression library from the salivary glands of Ornithodoros moubata and subsequently produced a self-assembled protein microarray using 480 randomly selected clones from that library. The reproducibility of the array, its representativeness of the tick salivary protein repertoire, and the functionality of the in situ expressed proteins have been checked, demonstrating that it is a suitable tool for the identification and functional characterization of soft tick salivary molecules that interact with host proteins. Several clones in the array were shown to bind to human recombinant P-selectin. One of them was a likely secreted tick phospholipase A2, which may represent a potential new ligand for P-selectin. As these salivary molecules are likely involved in blood meal acquisition through the modulation of the host immune and hemostatic responses, this new high-throughput tool could open new avenues for development of new therapeutic agents and control strategies against ticks and tick-borne pathogens.


Subject(s)
Arthropod Proteins/analysis , Ornithodoros/metabolism , Protein Array Analysis/methods , Salivary Glands/metabolism , Salivary Proteins and Peptides/analysis , Animals , Arthropod Proteins/metabolism , Cell-Free System , Female , Gene Library , High-Throughput Screening Assays , Humans , Male , Ornithodoros/genetics , P-Selectin/chemistry , Protein Binding , Protein Interaction Mapping , Proteomics/methods , Recombinant Proteins/metabolism , Reproducibility of Results , Salivary Glands/cytology , Salivary Proteins and Peptides/genetics , Salivary Proteins and Peptides/metabolism , Sequence Analysis, DNA
15.
Cir. Esp. (Ed. impr.) ; 77(3): 166-169, mar. 2005. ilus, tab
Article in Es | IBECS | ID: ibc-037747

ABSTRACT

Los feocromocitomas son tumores neuroendocrinospoco frecuentes, habitualmente sintomáticos y en la mayoría de los casos esporádicos, que se suelen localizar en la médula suprarrenal. Presentamos 2 casos de feocromocitoma extraadrenal no funcionan-te localizados en el retroperitoneo, aparentemente benignos, esporádicos y no asociados con otras neoplasias endocrinas ni enfermedades hereditarias. Empleamos ecografía, tomografía computarizada y resonancia magnética antes de la escisión completa del tumor por vía abdominal abierta, así como diversas técnicas inmunohistoquímicas para llegar al diagnóstico definitivo. Tras 8 y 14 meses de seguimiento no hay evidencia de recidiva de la enfermedad ni de aparición de otros síndromes. Como consecuencia de estos casos, llevamos a cabo una revisión bibliográfica centrándonos en los puntos más controvertidos (AU)


Pheochromocytomas are uncommon neuroendocrine tumors. They are usually symptomatic and sporadic and are generally located in the adrenal medulla. We present two cases of extra-adrenal nonfunctional pheochromocytomas located in the retro peritoneum. The tumors were apparently benign and sporadic and were not associated with other neoplasms or hereditary diseases. Ultrasonography, computed axial tomography and magnetic resonance imaging were performed before complete excision of the tumors using open abdominal surgery. Several immunohistochemical techniques were performed to reach the definitive diagnosis. After 8 and 14 months of follow-up, there is no evidence of recurrence or other syndromes. As a consequence of these two cases, we performed review of the literature on the topic, focussing on the most controversial areas (AU)


Subject(s)
Female , Adult , Middle Aged , Humans , Pheochromocytoma/diagnosis , Pheochromocytoma/surgery , Immunohistochemistry/methods , Paraganglioma/complications , Paraganglioma/diagnosis , Laparotomy/methods , Laparoscopy/methods , Abdomen/pathology , Abdomen , Sensitivity and Specificity , Paraganglioma/surgery
16.
Cir Esp ; 77(3): 166-9, 2005 Mar.
Article in Spanish | MEDLINE | ID: mdl-16420910

ABSTRACT

Pheochromocytomas are uncommon neuroendocrine tumors. They are usually symptomatic and sporadic and are generally located in the adrenal medulla. We present two cases of extra-adrenal nonfunctional pheochromocytomas located in the retroperitoneum. The tumors were apparently benign and sporadic and were not associated with other neoplasms or hereditary diseases. Ultrasonography, computed axial tomography and magnetic resonance imaging were performed before complete excision of the tumors using open abdominal surgery. Several immunohistochemical techniques were performed to reach the definitive diagnosis. After 8 and 14 months of follow-up, there is no evidence of recurrence or other syndromes. As a consequence of these two cases, we performed a review of the literature on the topic, focussing on the most controversial areas.


Subject(s)
Adrenal Gland Neoplasms/diagnostic imaging , Adrenal Gland Neoplasms/pathology , Pheochromocytoma/diagnostic imaging , Pheochromocytoma/pathology , Adrenal Gland Neoplasms/surgery , Adult , Aged , Female , Humans , Pheochromocytoma/surgery , Tomography, X-Ray Computed
17.
Arch. venez. farmacol. ter ; 6(4): 241-9, 1987. ilus, tab
Article in Spanish | LILACS | ID: lil-66652

ABSTRACT

Se seleccionaron 43 pacientes de ambos sexos con una edad comprendida entre 46 a 80 años, en cuatro Servicios de Reumatología integrados al Centro Nacional de Enfermedades Reumáticas del MSAS. Los pacientes presentaron inflamación, dolor en reposo, dolor a la presión, dolor a la movilización activa y pasiva de la articulación de una o ambas rodillas y limitación de la actividad física diaria. Todas las personas incluidas dieron su consentimiento para participar en este estudio. Excluyendo aquellos con úlcera péptica, embarazadas, madres amamantando y alérgicos a la aspirina u otros antiinflamatorios no esteroides (AINES). Los pacientes fueron tratados por 15 días con 20 mg de piroxicam en supositorios, y luego de una semana sin droga se comenzó la administración por 15 días más de diclofenac a la dosis diaria de 50 mg por vía oral. El dolor en reposo disminuyó en el 35.7% de los pacientes tratados con piroxicam en comparación con 8.4% por diclofenac. Hubo mejoría de la movilización pasiva de la rodilla con ambas drogas, sin diferencias significativas entre ellas. El dolor a la presión en la cara interna de la rodilla disminuyó significativamente más con el piroxicam que con el diclofenac. El dolor a la presión en la cara externa de la rodilla mejoró con ambos tratamientos, pero no hubo diferencia significativa entre los dos tipos de medicamento, igualmente ocurrió con el dolor al caminar y la actividad diaria del paciente. La velocidad para caminar 20 metros fue mejorada con los dos tratamientos en un rango de 0.1 a 2.5 segundos, sin embargo esta mejoría no fue estadísticamente significativa..


Subject(s)
Middle Aged , Humans , Male , Female , Anti-Inflammatory Agents/therapeutic use , Diclofenac/therapeutic use , Osteoarthritis/drug therapy , Piroxicam/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...