Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Immunol ; 207(7): 1891-1902, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34470857

RESUMO

Systemic duress, such as that elicited by sepsis, burns, or trauma, predisposes patients to secondary pneumonia, demanding better understanding of host pathways influencing this deleterious connection. These pre-existing circumstances are capable of triggering the hepatic acute-phase response (APR), which we previously demonstrated is essential for limiting susceptibility to secondary lung infections. To identify potential mechanisms underlying protection afforded by the lung-liver axis, our studies aimed to evaluate liver-dependent lung reprogramming when a systemic inflammatory challenge precedes pneumonia. Wild-type mice and APR-deficient littermate mice with hepatocyte-specific deletion of STAT3 (hepSTAT3-/-), a transcription factor necessary for full APR initiation, were challenged i.p. with LPS to induce endotoxemia. After 18 h, pneumonia was induced by intratracheal Escherichia coli instillation. Endotoxemia elicited significant transcriptional alterations in the lungs of wild-type and hepSTAT3-/- mice, with nearly 2000 differentially expressed genes between genotypes. The gene signatures revealed exaggerated immune activity in the lungs of hepSTAT3-/- mice, which were compromised in their capacity to launch additional cytokine responses to secondary infection. Proteomics revealed substantial liver-dependent modifications in the airspaces of pneumonic mice, implicating a network of dispatched liver-derived mediators influencing lung homeostasis. These results indicate that after systemic inflammation, liver acute-phase changes dramatically remodel the lungs, resulting in a modified landscape for any stimuli encountered thereafter. Based on the established vulnerability of hepSTAT3-/- mice to secondary lung infections, we believe that intact liver function is critical for maintaining the immunological responsiveness of the lungs.


Assuntos
Reação de Fase Aguda/imunologia , Coinfecção/imunologia , Fígado/metabolismo , Pulmão/patologia , Fator de Transcrição STAT3/metabolismo , Remodelação das Vias Aéreas , Animais , Células Cultivadas , Endotoxemia , Inflamação , Lipopolissacarídeos/metabolismo , Fígado/patologia , Camundongos , Camundongos Knockout , Proteômica , Fator de Transcrição STAT3/genética , Transcriptoma
2.
Cytometry A ; 101(11): 892-902, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-34854229

RESUMO

Recovery from pneumococcal (Spn) pneumonia induces development of tissue resident memory CD4+ TRM cells, BRM cells, and antibody secreting plasma cells in experienced lungs. These tissue resident lymphocytes confer protection against subsequent lethal challenge by serotype mismatched Spn (termed as heterotypic immunity). While traditional flow cytometry and gating strategies support premeditated identification of cells using a limited set of markers, discovery of novel tissue resident lymphocytes necessitates stable platforms that can handle larger sets of phenotypic markers and lends itself to unbiased clustering approaches. In this report, we leverage the power of full spectrum flow cytometry (FSFC) to develop a comprehensive panel of phenotypic markers that allows identification of multiple subsets of tissue resident lymphocytes in Spn-experienced murine lungs. Using Phenograph algorithm on this multidimensional data, we identify unforeseen heterogeneity in lung resident adaptive immune landscape which includes unexpected subsets of TRM and BRM cells. Further, using conventional gating strategy informed by our unsupervised clustering data, we confirm their presence exquisitely in Spn-experienced lungs as potentially relevant to heterotypic immunity and define CD73 as a highly expressed marker on TRM cells. Thus, our study emphasizes the utility of FSFC for confirmatory and discovery studies relating to tissue resident adaptive immunity.


Assuntos
Pneumonia Pneumocócica , Camundongos , Animais , Memória Imunológica , Pulmão , Linfócitos T CD8-Positivos , Linfócitos
3.
Traffic ; 17(3): 289-300, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26602861

RESUMO

Ligand stimulation promotes downregulation of RTKs, a mechanism by which RTKs, through the ubiquitination pathway are removed from the cell surface, causing a temporary termination of RTK signaling. The molecular mechanisms governing RTK trafficking and maturation in the endoplasmic reticulum (ER)/Golgi compartments are poorly understood. Vascular endothelial growth factor receptor-2 (VEGFR-2) is a prototypic RTK that plays a critical role in physiologic and pathologic angiogenesis. Here we demonstrate that Ring Finger Protein 121 (RNF121), an ER ubiquitin E3 ligase, is expressed in endothelial cells and regulates maturation of VEGFR-2. RNF121 recognizes newly synthesized VEGFR-2 in the ER and controls its trafficking and maturation. Over-expression of RNF121 promoted ubiquitination of VEGFR-2, inhibited its maturation and resulted a significantly reduced VEGFR-2 presence at the cell surface. Conversely, the shRNA-mediated knockdown of RNF121 in primary endothelial cells reduced VEGFR-2 ubiquitination and increased its cell surface level. The RING Finger domain of RNF121 is required for its activity toward VEGFR-2, as its deletion significantly reduced the effect of RNF121 on VEGFR-2. Additionally, RNF121 inhibited VEGF-induced endothelial cell proliferation and angiogenesis. Taken together, these data identify RNF121 as a key determinant of angiogenic signaling that restricts VEGFR-2 cell surface presence and its angiogenic signaling.


Assuntos
Membrana Celular/metabolismo , Proteínas de Membrana/metabolismo , Transdução de Sinais , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Proliferação de Células , Retículo Endoplasmático/metabolismo , Células HEK293 , Células HT29 , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/fisiologia , Humanos , Proteínas de Membrana/genética , Transporte Proteico , Suínos , Ubiquitinação , Fator A de Crescimento do Endotélio Vascular/metabolismo
4.
Am J Pathol ; 185(10): 2757-67, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26342724

RESUMO

Oxidative damage to renal tubular epithelial cells is a fundamental pathogenic mechanism implicated in both acute kidney injury and chronic kidney diseases. Because epithelial cell survival influences the outcome of acute kidney injury and chronic kidney diseases, identifying its molecular regulators could provide new insight into pathobiology and possible new therapeutic strategies for these diseases. We have identified transmembrane and immunoglobulin domain-containing 1 (TMIGD1) as a novel adhesion molecule, which is highly conserved in humans and other species. TMIGD1 is expressed in renal tubular epithelial cells and promotes cell survival. The extracellular domain of TMIGD1 contains two putative immunoglobulin domains and mediates self-dimerization. Our data suggest that TMIGD1 regulates transepithelial electric resistance and permeability of renal epithelial cells. TMIGD1 controls cell migration, cell morphology, and protects renal epithelial cells from oxidative- and nutrient-deprivation-induced cell injury. Hydrogen peroxide-induced oxidative cell injury downregulates TMIGD1 expression and targets it for ubiquitination. Moreover, TMIGD1 expression is significantly affected in both acute kidney injury and in deoxy-corticosterone acetate and sodium chloride (deoxy-corticosterone acetate salt)-induced chronic hypertensive kidney disease mouse models. Taken together, we have identified TMIGD1 as a novel cell adhesion molecule expressed in kidney epithelial cells that protects kidney epithelial cells from oxidative cell injury to promote cell survival.


Assuntos
Injúria Renal Aguda/metabolismo , Células Epiteliais/metabolismo , Glicoproteínas de Membrana/metabolismo , Estresse Oxidativo/fisiologia , Injúria Renal Aguda/patologia , Animais , Moléculas de Adesão Celular/metabolismo , Humanos , Peróxido de Hidrogênio/metabolismo , Imunoglobulinas/metabolismo , Túbulos Renais Proximais/metabolismo , Túbulos Renais Proximais/patologia , Camundongos da Linhagem 129 , RNA Mensageiro/metabolismo
5.
Front Immunol ; 15: 1382638, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38715601

RESUMO

Recovery from respiratory pneumococcal infections generates lung-localized protection against heterotypic bacteria, mediated by resident memory lymphocytes. Optimal protection in mice requires re-exposure to pneumococcus within days of initial infection. Serial surface marker phenotyping of B cell populations in a model of pneumococcal heterotypic immunity revealed that bacterial re-exposure stimulates the immediate accumulation of dynamic and heterogeneous populations of B cells in the lung, and is essential for the establishment of lung resident memory B (BRM) cells. The B cells in the early wave were activated, proliferating locally, and associated with both CD4+ T cells and CXCL13. Antagonist- and antibody-mediated interventions were implemented during this early timeframe to demonstrate that lymphocyte recirculation, CD4+ cells, and CD40 ligand (CD40L) signaling were all needed for lung BRM cell establishment, whereas CXCL13 signaling was not. While most prominent as aggregates in the loose connective tissue of bronchovascular bundles, morphometry and live lung imaging analyses showed that lung BRM cells were equally numerous as single cells dispersed throughout the alveolar septae. We propose that CD40L signaling from antigen-stimulated CD4+ T cells in the infected lung is critical to establishment of local BRM cells, which subsequently protect the airways and parenchyma against future potential infections.


Assuntos
Linfócitos T CD4-Positivos , Ligante de CD40 , Pulmão , Células B de Memória , Streptococcus pneumoniae , Animais , Camundongos , Linfócitos T CD4-Positivos/imunologia , Ligante de CD40/metabolismo , Ligante de CD40/imunologia , Quimiocina CXCL13/metabolismo , Modelos Animais de Doenças , Memória Imunológica , Pulmão/imunologia , Células B de Memória/imunologia , Células B de Memória/metabolismo , Camundongos Endogâmicos C57BL , Infecções Pneumocócicas/imunologia , Transdução de Sinais , Streptococcus pneumoniae/imunologia
6.
Mucosal Immunol ; 16(5): 699-710, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37604254

RESUMO

Streptococcus pneumoniae is the most common etiology of bacterial pneumonia, one of the leading causes of death in children and the elderly worldwide. During non-lethal infections with S. pneumoniae, lymphocytes accumulate in the lungs and protect against reinfection with serotype-mismatched strains. Cluster of differentiation CD4+ resident memory T (TRM) cells are known to be crucial for this protection, but the diversity of lung CD4+ TRM cells has yet to be fully delineated. We aimed to identify unique subsets and their contributions to lung immunity. After recovery from pneumococcal infections, we identified a distinct subset of CD4+ T cells defined by the phenotype CD11ahiCD69+GL7+ in mouse lungs. Phenotypic analyses for markers of lymphocyte memory and residence demonstrated that GL7+ T cells are a subset of CD4+ TRM cells. Functional studies revealed that unlike GL7- TRM subsets that were mostly (RAR-related Orphan Receptor gamma T) RORγT+, GL7+ TRM cells exhibited higher levels of (T-box expressed in T cells) T-bet and Gata-3, corresponding with increased synthesis of interferon-γ, interleukin-13, and interleukin-5, inherent to both T helper 1 (TH1) and TH2 functions. Thus, we propose that these cells provide novel contributions during pneumococcal pneumonia, serving as important determinants of lung immunity.


Assuntos
Pulmão , Streptococcus pneumoniae , Idoso , Animais , Criança , Humanos , Camundongos , Linfócitos T CD4-Positivos , Memória Imunológica , Ligantes , Linfócitos T
7.
JCI Insight ; 7(5)2022 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-35133985

RESUMO

Recovery from pneumococcal pneumonia remodels the pool of alveolar macrophages so that they exhibit new surface marker profiles, transcriptomes, metabolomes, and responses to infection. Mechanisms mediating alveolar macrophage phenotypes after pneumococcal pneumonia have not been delineated. IFN-γ and its receptor on alveolar macrophages were essential for certain, but not all, aspects of the remodeled alveolar macrophage phenotype. IFN-γ was produced by CD4+ T cells plus other cells, and CD4+ cell depletion did not prevent alveolar macrophage remodeling. In mice infected or recovering from pneumococcus, monocytes were recruited to the lungs, and the monocyte-derived macrophages developed characteristics of alveolar macrophages. CCR2 mediated the early monocyte recruitment but was not essential to the development of the remodeled alveolar macrophage phenotype. Lineage tracing demonstrated that recovery from pneumococcal pneumonias converted the pool of alveolar macrophages from being primarily of embryonic origin to being primarily of adult hematopoietic stem cell origin. Alveolar macrophages of either origin demonstrated similar remodeled phenotypes, suggesting that ontogeny did not dictate phenotype. Our data reveal that the remodeled alveolar macrophage phenotype in lungs recovered from pneumococcal pneumonia results from a combination of new recruitment plus training of both the original cells and the new recruits.


Assuntos
Macrófagos Alveolares , Pneumonia Pneumocócica , Animais , Pulmão , Macrófagos , Camundongos , Monócitos
8.
JCI Insight ; 7(23)2022 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-36264633

RESUMO

Identifying host factors that contribute to pneumonia incidence and severity are of utmost importance to guiding the development of more effective therapies. Lectin-like oxidized low-density lipoprotein receptor 1 (LOX-1, encoded by OLR1) is a scavenger receptor known to promote vascular injury and inflammation, but whether and how LOX-1 functions in the lung are unknown. Here, we provide evidence of substantial accumulation of LOX-1 in the lungs of patients with acute respiratory distress syndrome and in mice with pneumonia. Unlike previously described injurious contributions of LOX-1, we found that LOX-1 is uniquely protective in the pulmonary airspaces, limiting proteinaceous edema and inflammation. We also identified alveolar macrophages and recruited neutrophils as 2 prominent sites of LOX-1 expression in the lungs, whereby macrophages are capable of further induction during pneumonia and neutrophils exhibit a rapid, but heterogenous, elevation of LOX-1 in the infected lung. Blockade of LOX-1 led to dysregulated immune signaling in alveolar macrophages, marked by alterations in activation markers and a concomitant elevation of inflammatory gene networks. However, bone marrow chimeras also suggested a prominent role for neutrophils in LOX-1-mediated lung protection, further supported by LOX-1+ neutrophils exhibiting transcriptional changes consistent with reparative processes. Taken together, this work establishes LOX-1 as a tissue-protective factor in the lungs during pneumonia, possibly mediated by its influence on immune signaling in alveolar macrophages and LOX-1+ airspace neutrophils.


Assuntos
Lesão Pulmonar , Pneumonia , Receptores Depuradores Classe E , Animais , Camundongos , Receptores Depuradores Classe E/genética
9.
J Clin Invest ; 131(11)2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-34060477

RESUMO

Lung-resident memory B cells (BRM cells) are elicited after influenza infections of mice, but connections to other pathogens and hosts - as well as their functional significance - have yet to be determined. We postulate that BRM cells are core components of lung immunity. To test this, we examined whether lung BRM cells are elicited by the respiratory pathogen pneumococcus, are present in humans, and are important in pneumonia defense. Lungs of mice that had recovered from pneumococcal infections did not contain organized tertiary lymphoid organs, but did have plasma cells and noncirculating memory B cells. The latter expressed distinctive surface markers (including CD69, PD-L2, CD80, and CD73) and were poised to secrete antibodies upon stimulation. Human lungs also contained B cells with a resident memory phenotype. In mice recovered from pneumococcal pneumonia, depletion of PD-L2+ B cells, including lung BRM cells, diminished bacterial clearance and the level of pneumococcus-reactive antibodies in the lung. These data define lung BRM cells as a common feature of pathogen-experienced lungs and provide direct evidence of a role for these cells in pulmonary antibacterial immunity.


Assuntos
Linfócitos B/imunologia , Memória Imunológica , Pulmão/imunologia , Pneumonia Pneumocócica/imunologia , Pneumonia Pneumocócica/prevenção & controle , Streptococcus pneumoniae/imunologia , Animais , Antígenos de Diferenciação/imunologia , Linfócitos B/patologia , Humanos , Pulmão/microbiologia , Pulmão/patologia , Camundongos , Camundongos Transgênicos , Pneumonia Pneumocócica/microbiologia , Pneumonia Pneumocócica/patologia
10.
Nat Commun ; 12(1): 5834, 2021 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-34611166

RESUMO

Barrier tissues are populated by functionally plastic CD4+ resident memory T (TRM) cells. Whether the barrier epithelium regulates CD4+ TRM cell locations, plasticity and activities remains unclear. Here we report that lung epithelial cells, including distinct surfactant protein C (SPC)lowMHChigh epithelial cells, function as anatomically-segregated and temporally-dynamic antigen presenting cells. In vivo ablation of lung epithelial MHC-II results in altered localization of CD4+ TRM cells. Recurrent encounters with cognate antigen in the absence of epithelial MHC-II leads CD4+ TRM cells to co-express several classically antagonistic lineage-defining transcription factors, changes their cytokine profiles, and results in dysregulated barrier immunity. In addition, lung epithelial MHC-II is needed for surface expression of PD-L1, which engages its ligand PD-1 to constrain lung CD4+ TRM cell phenotypes. Thus, we establish epithelial antigen presentation as a critical regulator of CD4+ TRM cell function and identify epithelial-CD4+ TRM cell immune interactions as core elements of barrier immunity.


Assuntos
Apresentação de Antígeno/fisiologia , Células Epiteliais/metabolismo , Pulmão/citologia , Animais , Linfócitos T CD4-Positivos/metabolismo , Citometria de Fluxo , Imunofluorescência , Leucócitos/citologia , Leucócitos/metabolismo , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão , Reação em Cadeia da Polimerase em Tempo Real
11.
Front Immunol ; 11: 554, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32300347

RESUMO

Cyclic di-AMP (c-di-AMP) is an important signaling molecule for pneumococci, and as a uniquely prokaryotic product it can be recognized by mammalian cells as a danger signal that triggers innate immunity. Roles of c-di-AMP in directing host responses during pneumococcal infection are only beginning to be defined. We hypothesized that pneumococci with defective c-di-AMP catabolism due to phosphodiesterase deletions could illuminate roles of c-di-AMP in mediating host responses to pneumococcal infection. Pneumococci deficient in phosphodiesterase 2 (Pde2) stimulated a rapid induction of interferon ß (IFNß) expression that was exaggerated in comparison to that induced by wild type (WT) bacteria or bacteria deficient in phosphodiesterase 1. This IFNß burst was elicited in mouse and human macrophage-like cell lines as well as in primary alveolar macrophages collected from mice with pneumococcal pneumonia. Macrophage hyperactivation by Pde2-deficient pneumococci led to rapid cell death. STING and cGAS were essential for the excessive IFNß induction, which also required phagocytosis of bacteria and triggered the phosphorylation of IRF3 and IRF7 transcription factors. The select effects of Pde2 deletion were products of a unique role of this enzyme in c-di-AMP catabolism when pneumococci were grown on solid substrate conditions designed to enhance virulence. Because pneumococci with elevated c-di-AMP drive aberrant innate immune responses from macrophages involving hyperactivation of STING, excessive IFNß expression, and rapid cytotoxicity, we surmise that c-di-AMP is pivotal for directing innate immunity and host-pathogen interactions during pneumococcal pneumonia.


Assuntos
Proteínas de Bactérias/imunologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 2/imunologia , Fosfatos de Dinucleosídeos/imunologia , Imunidade Inata/imunologia , Macrófagos/imunologia , Streptococcus pneumoniae/imunologia , Animais , Proteínas de Bactérias/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 2/metabolismo , Fosfatos de Dinucleosídeos/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Pneumonia Pneumocócica/imunologia , Células RAW 264.7
12.
Mucosal Immunol ; 13(2): 334-343, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31748706

RESUMO

Previous pneumococcal experience establishes lung-resident IL-17A-producing CD4+ memory TRM cells that accelerate neutrophil recruitment against heterotypic pneumococci. Herein, we unravel a novel crosstalk between CD4+ TRM cells and lung epithelial cells underlying this protective immunity. Depletion of CD4+ cells in pneumococcus-experienced mice diminished CXCL5 (but not CXCL1 or CXCL2) and downstream neutrophil accumulation in the lungs. Epithelial cells from experienced lungs exhibited elevated mRNA for CXCL5 but not other epithelial products such as GM-CSF or CCL20, suggesting a skewing by CD4+ TRM cells. Genome-wide expression analyses revealed a significant remodeling of the epithelial transcriptome of infected lungs due to infection history, ~80% of which was CD4+ cell-dependent. The CD4+ TRM cell product IL-17A stabilized CXCL5 but not GM-CSF or CCL20 mRNA in cultured lung epithelial cells, implicating posttranscriptional regulation as a mechanism for altered epithelial responses. These results suggest that epithelial cells in experienced lungs are effectively different, owing to their communication with TRM cells. Our study highlights the role of tissue-resident adaptive immune cells in fine-tuning epithelial functions to hasten innate immune responses and optimize defense in experienced lungs, a concept that may apply broadly to mucosal immunology.


Assuntos
Pulmão/imunologia , Neutrófilos/imunologia , Pneumonia Pneumocócica/imunologia , Mucosa Respiratória/fisiologia , Streptococcus pneumoniae/fisiologia , Células Th17/imunologia , Remodelação das Vias Aéreas , Animais , Comunicação Celular , Quimiocina CXCL5/genética , Quimiocina CXCL5/metabolismo , Regulação da Expressão Gênica , Humanos , Doenças do Sistema Imunitário , Imunidade Inata , Memória Imunológica , Transtornos Leucocíticos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
13.
JCI Insight ; 5(4)2020 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-31990682

RESUMO

Community-acquired pneumonia is a widespread disease with significant morbidity and mortality. Alveolar macrophages are tissue-resident lung cells that play a crucial role in innate immunity against bacteria that cause pneumonia. We hypothesized that alveolar macrophages display adaptive characteristics after resolution of bacterial pneumonia. We studied mice 1 to 6 months after self-limiting lung infections with Streptococcus pneumoniae, the most common cause of bacterial pneumonia. Alveolar macrophages, but not other myeloid cells, recovered from the lung showed long-term modifications of their surface marker phenotype. The remodeling of alveolar macrophages was (a) long-lasting (still observed 6 months after infection), (b) regionally localized (observed only in the affected lobe after lobar pneumonia), and (c) associated with macrophage-dependent enhanced protection against another pneumococcal serotype. Metabolomic and transcriptomic profiling revealed that alveolar macrophages of mice that recovered from pneumonia had new baseline activities and altered responses to infection that better resembled those of adult humans. The enhanced lung protection after mild and self-limiting bacterial respiratory infections includes a profound remodeling of the alveolar macrophage pool that is long-lasting; compartmentalized; and manifest across surface receptors, metabolites, and both resting and stimulated transcriptomes.


Assuntos
Macrófagos Alveolares/imunologia , Pneumonia Pneumocócica/imunologia , Animais , Diferenciação Celular , Modelos Animais de Doenças , Imunidade Inata , Pulmão/imunologia , Camundongos , Células Mieloides/citologia , Células Mieloides/imunologia
14.
Int. j. morphol ; 35(4): 1276-1279, Dec. 2017. graf
Artigo em Inglês | LILACS | ID: biblio-893127

RESUMO

SUMMARY: Variation in the morphology of the extensor pollicis longus muscle is very rare. The muscle is considered as one of the deep group of dorsal forearm muscles. It passes through the dorsal radiocarpal ligament in a detached compartment. It inserts at the base of the distal phalanx of the thumb. The muscle might have double tendons where they run either in one, or in separate compartments. In this study, we report an extremely rare condition for a 38-year-old male where some of the tendon fibers split at the base of the proximal phalanx and the two tendons insert at the sides of the base of the distal phalanx. We also noticed a high range of hyperextension where the action was painless and showed no sign of discomfort. We believe that the way the tendons get inserted might have a role in this wide range of extension.


RESUMEN: La variación en la morfología del músculo extensor largo del pulgar es muy rara. Es considerado perteneciente al grupo profundo de los músculos del dorso del antebrazo. Su tendón pasa a través del ligamento radiocarpiano dorsal en un compartimiento separado. Se inserta en la base de la falange distal del pulgar. El músculo puede tener tendones dobles que pasan a través de un solo compartimiento, o en compartimentos separados, del ligamento radiocarpiano dorsal. En este estudio se reporta una variación extremadamente rara en un hombre de 38 años de edad, donde algunas de las fibras del tendón se dividen en la base de la falange proximal y los dos tendones se insertaban a los lados de la base de la falange distal del pulgar. También observamos un alto grado de hiperextensión siendo la acción indolora y no mostrando ningún signo de malestar. Creemos que la forma en que los tendones se insertan podrían tener un papel en este amplio rango de extensión.


Assuntos
Humanos , Masculino , Adulto , Articulações dos Dedos/anormalidades , Músculo Esquelético/anormalidades , Tendões/anormalidades , Variação Anatômica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA