Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Clin Med (Lond) ; 24(3): 100203, 2024 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-38643827

RESUMO

Megakaryocytes are large multilobulated precursor cells which usually reside within the bone marrow and give rise to platelets. There have been rare occurrences where they have been found in peripheral blood and extramedullary tissues in conditions where the underlying mechanisms of the bone marrow have been affected. This case report discusses an unusual presentation of a man with myelofibrosis who was found to have megakaryocytes in his ascitic fluid. We have highlighted the images showing utility of combination of traditional staining methods and immunohistochemistry in combating this diagnostic dilemma.

2.
Mol Cancer Ther ; 22(11): 1319-1331, 2023 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-37486980

RESUMO

Chimeric antigen receptor (CAR) T-cell immunotherapies for solid tumors face critical challenges such as heterogeneous antigen expression. We characterized stage-specific embryonic antigen-4 (SSEA-4) cell-surface glycolipid as a target for CAR T-cell therapy. SSEA-4 is mainly expressed during embryogenesis but is also found in several cancer types making it an attractive tumor-associated antigen. Anti-SSEA-4 CAR-T cells were generated and assessed preclinically in vitro and in vivo for antitumor response and safety. SSEA-4 CAR-T cells effectively eliminated SSEA-4-positive cells in all the tested cancer cell lines, whereas SSEA-4-negative cells lines were not targeted. In vivo efficacy and safety studies using NSG mice and the high-grade serous ovarian cancer cell line OVCAR4 demonstrated a remarkable and specific antitumor response at all the CAR T-cell doses used. At high T-cell doses, CAR T cell-treated mice showed signs of health deterioration after a follow-up period. However, the severity of toxicity was reduced with a delayed onset when lower CAR T-cell doses were used. Our data demonstrate the efficacy of anti-SSEA-4 CAR T-cell therapy; however, safety strategies, such as dose-limiting and/or equipping CAR-T cells with combinatorial antigen recognition should be implemented for its potential clinical translation.


Assuntos
Carcinoma , Neoplasias Ovarianas , Receptores de Antígenos Quiméricos , Humanos , Feminino , Animais , Camundongos , Glicoesfingolipídeos/metabolismo , Linhagem Celular Tumoral , Neoplasias Ovarianas/metabolismo , Imunoterapia Adotiva , Linfócitos T , Carcinoma Epitelial do Ovário/metabolismo , Carcinoma/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
3.
J Trauma Acute Care Surg ; 95(4): 481-489, 2023 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-37249511

RESUMO

BACKGROUND: Cardiac dysfunction (CD) has emerged as a key contributor to delayed organ failure and late mortality in patients surviving the initial traumatic hemorrhagic response. Inflammatory processes are implicated in the initial stages of this CD; however, downstream pathways leading to a characteristic rapid fall in stroke volume and cardiac output are not yet fully defined. Currently, no cardioprotective treatments are available. We investigated the role of myocardial oxidative stress in the pathogenesis of CD associated to traumatic hemorrhagic injury, and its related metabolomic profile. METHODS: Ex vivo tissue from a 3-hour murine model of pressure-controlled trauma hemorrhagic shock (THS) was analyzed. Animals were randomized to echocardiography-guided crystalloid fluid resuscitation or a control group (sham: cannulation and anesthesia only, or naïve: no intervention). Trauma hemorrhagic shock and naïve samples were assessed by immunohistochemistry for nuclear 8-hydroxy-2'-deoxyguanosine expression as a marker of oxidative stress. Metabolomic analysis of THS and sham group tissue was performed by LC-MS. RESULTS: 8-Hydroxy-2'-deoxyguanosine expression across the myocardium was significantly higher following THS injury compared to naïve group (33.01 ± 14.40% vs. 15.08 ± 3.96%, p < 0.05). Trauma hemorrhagic shock injury significantly increased lysine ( p = 0.022), and decreased aconitate ( p = 0.016) and glutamate ( p = 0.047) in the myocardium, indicating activation of a catabolic metabolism and oxidative stress response. CONCLUSION: We confirm the acute development of oxidative stress lesions and altered cardiac energy metabolism following traumatic hemorrhage injury, providing insight into the relationship between inflammatory damage and impaired cardiac contractility. These findings may provide targets for development of novel cardioprotective therapeutics aiming to decrease late mortality from trauma.


Assuntos
Lesões por Esmagamento , Choque Hemorrágico , Animais , Humanos , Camundongos , 8-Hidroxi-2'-Desoxiguanosina , Coração , Hemorragia/etiologia , Hemorragia/terapia , Miocárdio , Choque Hemorrágico/terapia
4.
Nanomaterials (Basel) ; 11(7)2021 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-34199005

RESUMO

The use of nanomaterials incorporated into plastic products is increasing steadily. By using nano-scaled filling materials, thermoplastics, such as polyethylene (PE), take advantage of the unique properties of nanomaterials (NM). The life cycle of these so-called nanocomposites (NC) usually ends with energetic recovery. However, the toxicity of these aerosols, which may consist of released NM as well as combustion-generated volatile compounds, is not fully understood. Within this study, model nanocomposites consisting of a PE matrix and nano-scaled filling material (TiO2, CuO, carbon nano tubes (CNT)) were produced and subsequently incinerated using a lab-scale model burner. The combustion-generated aerosols were characterized with regard to particle release as well as compound composition. Subsequently, A549 cells and a reconstituted 3D lung cell culture model (MucilAir™, Epithelix) were exposed for 4 h to the respective aerosols. This approach enabled the parallel application of a complete aerosol, an aerosol under conditions of enhanced particle deposition using high voltage, and a filtered aerosol resulting in the sole gaseous phase. After 20 h post-incubation, cytotoxicity, inflammatory response (IL-8), transcriptional toxicity profiling, and genotoxicity were determined. Only the exposure toward combustion aerosols originated from PE-based materials induced cytotoxicity, genotoxicity, and transcriptional alterations in both cell models. In contrast, an inflammatory response in A549 cells was more evident after exposure toward aerosols of nano-scaled filler combustion, whereas the thermal decomposition of PE-based materials revealed an impaired IL-8 secretion. MucilAir™ tissue showed a pronounced inflammatory response after exposure to either combustion aerosols, except for nanocomposite combustion. In conclusion, this study supports the present knowledge on the release of nanomaterials after incineration of nano-enabled thermoplastics. Since in the case of PE-based combustion aerosols no major differences were evident between exposure to the complete aerosol and to the gaseous phase, adverse cellular effects could be deduced to the volatile organic compounds that are generated during incomplete combustion of NC.

5.
Int J Mol Sci ; 22(9)2021 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-34068728

RESUMO

To mimic more realistic lung tissue conditions, co-cultures of epithelial and immune cells are one comparatively easy-to-use option. To reveal the impact of immune cells on the mode of action (MoA) of CuO nanoparticles (NP) on epithelial cells, A549 cells as a model for epithelial cells have been cultured with or without differentiated THP-1 cells, as a model for macrophages. After 24 h of submerged incubation, cytotoxicity and transcriptional toxicity profiles were obtained and compared between the cell culture systems. Dose-dependent cytotoxicity was apparent starting from 8.0 µg/cm2 CuO NP. With regard to gene expression profiles, no differences between the cell models were observed concerning metal homeostasis, oxidative stress, and DNA damage, confirming the known MoA of CuO NP, i.e., endocytotic particle uptake, intracellular particle dissolution within lysosomes with subsequent metal ion deliberation, increased oxidative stress, and genotoxicity. However, applying a co-culture of epithelial and macrophage-like cells, CuO NP additionally provoked a pro-inflammatory response involving NLRP3 inflammasome and pro-inflammatory transcription factor activation. This study demonstrates that the application of this easy-to-use advanced in vitro model is able to extend the detection of cellular effects provoked by nanomaterials by an immunological response and emphasizes the use of such models to address a more comprehensive MoA.


Assuntos
Epitélio/efeitos dos fármacos , Nanopartículas Metálicas/química , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Estresse Oxidativo/genética , Transcrição Gênica/efeitos dos fármacos , Células A549 , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Técnicas de Cocultura , Cobre/química , Cobre/farmacologia , Dano ao DNA/efeitos dos fármacos , Endocitose/efeitos dos fármacos , Humanos , Pulmão/efeitos dos fármacos , Pulmão/patologia , Macrófagos/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo
6.
Nanomaterials (Basel) ; 12(1)2021 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-35010097

RESUMO

While the toxicity of metal-based nanoparticles (NP) has been investigated in an increasing number of studies, little is known about metal-based fibrous materials, so-called nanowires (NWs). Within the present study, the physico-chemical properties of particulate and fibrous nanomaterials based on Cu, CuO, Ni, and Ag as well as TiO2 and CeO2 NP were characterized and compared with respect to abiotic metal ion release in different physiologically relevant media as well as acellular reactivity. While none of the materials was soluble at neutral pH in artificial alveolar fluid (AAF), Cu, CuO, and Ni-based materials displayed distinct dissolution under the acidic conditions found in artificial lysosomal fluids (ALF and PSF). Subsequently, four different cell lines were applied to compare cytotoxicity as well as intracellular metal ion release in the cytoplasm and nucleus. Both cytotoxicity and bioavailability reflected the acellular dissolution rates in physiological lysosomal media (pH 4.5); only Ag-based materials showed no or very low acellular solubility, but pronounced intracellular bioavailability and cytotoxicity, leading to particularly high concentrations in the nucleus. In conclusion, in spite of some quantitative differences, the intracellular bioavailability as well as toxicity is mostly driven by the respective metal and is less modulated by the shape of the respective NP or NW.

7.
Chem Res Toxicol ; 33(5): 1237-1249, 2020 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-32285662

RESUMO

To assess the toxicity of nanomaterials, most in vitro studies have been performed under submerged conditions, which do not reflect physiological conditions upon inhalation. An air-liquid interface (ALI) exposure may provide more reliable data on dosimetry and prevent interactions with cell culture media components. Therefore, an ALI exposure was combined with a high-throughput RT-qPCR approach to evaluate the toxicological potential of CuO and TiO2 nanoparticles (NP) in A549 cells. While TiO2 NP did not show any cytotoxicity or other effects compromising genomic stability up to 25.8 µg/cm2, CuO NP revealed a dose-dependent cytotoxicity, starting at 4.9 µg/cm2. Furthermore, CuO NP altered distinct gene expression patterns indicative for disturbed metal homeostasis, stress response, and DNA damage induction. Thus, induction of metal homeostasis associated genes (MT1X, MT2A) at 0.4 µg/cm2 and higher suggested uptake and intracellular dissolution of CuO NP, which was verified by a dose-dependent increase in intracellular copper concentration. Starting at 4.9 µg/cm2, oxidative stress markers (HMOX1, HSPA1A) were induced dose-dependently, supported by elevated ROS levels. Furthermore, a dose-dependent induction of genes associated with DNA damage response (DDIT3, GADD45A) was observed, in concordance with an increase in DNA strand breaks. Finally, transcriptional data suggested the induction of apoptosis at high doses, while flow cytometric analysis revealed increased numbers of either late apoptotic or necrotic cells and clearly necrotic cells at the highest concentrations. Thus, an ALI cell culture system was successfully combined with a comprehensive high-throughput RT-qPCR system, allowing the quantification of NP deposition and their impact on genomic stability. For CuO NP, in principle the data confirm observations made under submerged conditions with respect to intracellular copper ion release, as well as oxidative and genotoxic stress response. However, the results derived from ALI exposure allow the assessment of dose-response-relationships as well as the comparison of relative toxic potencies of different NP.


Assuntos
Cobre/toxicidade , Perfilação da Expressão Gênica , Nanopartículas Metálicas/química , Nanopartículas Metálicas/toxicidade , Titânio/toxicidade , Células A549 , Ar , Apoptose/efeitos dos fármacos , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/genética , Cobre/química , Dano ao DNA , Relação Dose-Resposta a Droga , Proteínas de Choque Térmico HSP70/antagonistas & inibidores , Proteínas de Choque Térmico HSP70/genética , Heme Oxigenase-1/antagonistas & inibidores , Heme Oxigenase-1/genética , Humanos , Metalotioneína/antagonistas & inibidores , Metalotioneína/genética , Estresse Oxidativo/efeitos dos fármacos , Tamanho da Partícula , Reação em Cadeia da Polimerase em Tempo Real , Titânio/química , Fator de Transcrição CHOP/antagonistas & inibidores , Fator de Transcrição CHOP/genética , Células Tumorais Cultivadas , Água/química
8.
Front Immunol ; 10: 2774, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31866998

RESUMO

Cardiac dysfunction (CD) importantly contributes to mortality in trauma patients, who survive their initial injuries following successful hemostatic resuscitation. This poor outcome has been correlated with elevated biomarkers of myocardial injury, but the pathophysiology triggering this CD remains unknown. We investigated the pathophysiology of acute CD after trauma using a mouse model of trauma hemorrhage shock (THS)-induced CD with echocardiographic guidance of fluid resuscitation, to assess the THS impact on myocardial integrity and function. Mice were subjected to trauma (soft tissue and bone fracture) and different degrees of hemorrhage severity (pressure controlled ~MABP < 35 mmHg or <65 mmHg) for 1 h, to characterize the acute impact on cardiac function. In a second study, mice were subjected to trauma and hemorrhage (MABP < 35 mmHg) for 1 h, then underwent two echocardiographic-guided resuscitations to baseline stroke volume at 60 and 120 min, and were monitored up to 180 min to study the longer impact of THS following resuscitation. Naïve and sham animals were used as controls. At 60 min post-THS injury, animals showed a lower cardiac output (CO) and stroke volume (SV) and an early rise of heart fatty acid-binding protein (H-FABP = 167 ± 38 ng/ml; 90% increase from shams, 3.54 ± 3.06 ng/ml), when subjected to severe hemorrhage and injury. Despite resuscitation, these animals maintained lower CO (6 ml/min vs. 23 ml/min), lower SV (10 µl vs. 46 µl; both ~75% decreased), and higher H-FABP (levels (340 ± 115 ng/ml vs. 10.3 ± 0.2 ng/ml; all THS vs. shams, P < 0.001) at 180 min post-THS injury. Histopathological and flow-cytometry analysis of the heart confirmed an influx of circulatory leukocytes, compared to non-injured hearts. Myocardial injury was supported by an increase of troponin I and h-FABP and the widespread ultrastructural disorganization of the morphology of sarcomeres and mitochondria. DNA fragmentation and chromatin condensation driven by leakage of apoptosis-inducing factor (AIF) may suggest a mitochondria-driven progressive cell death. THS modeling in the mouse results in cardiomyocyte damage and reduced myocardial function, which mimics the cardiac dysfunction seen in trauma patients. This CD model may, therefore, provide further understanding to the mechanisms underlying CD and act as a tool for developing cardioprotective therapeutics to improve survival after injury.


Assuntos
Hemorragia/complicações , Hemorragia/etiologia , Modelos Cardiovasculares , Disfunção Ventricular/etiologia , Disfunção Ventricular/fisiopatologia , Ferimentos e Lesões/complicações , Animais , Biomarcadores , Modelos Animais de Doenças , Coração/fisiopatologia , Hemodinâmica , Humanos , Masculino , Camundongos , Miocárdio/metabolismo , Choque Hemorrágico/etiologia
9.
Histopathology ; 75(6): 813-824, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31310679

RESUMO

AIMS: Lynch syndrome (LS) is associated with an increased risk of developing endometrial carcinoma (EC) and ovarian carcinoma (OC). There is considerable variability in current practices and opinions related to screening of newly diagnosed patients with EC/OC for LS. An online survey was undertaken to explore the extent of these differences. METHODS AND RESULTS: An online questionnaire was developed by a panel of experts and sent to all members of the British Association of Gynaecological Pathologists (BAGP) and the International Society of Gynecological Pathologists (ISGyP). Anonymised results were received and analysed. Thirty-six BAGP and 44 ISGyP members completed the survey. More than 90% of respondents were aware of the association of LS with both EC and OC, but 34% were not aware of specific guidelines for LS screening. Seventy-one per cent of respondents agreed that universal screening for LS should be carried out in all newly diagnosed EC cases, with immunohistochemistry (IHC) alone as the preferred approach. Only 36% of respondents currently performed IHC or microsatellite instability testing on all newly diagnosed EC cases, with most of the remaining respondents practising selective screening, based on clinical or pathological features or both. A significant minority of respondents (35%) believed that patient consent was required before performance of mismatch repair (MMR) protein IHC. Almost all respondents favoured the use of standardised terminology for reporting MMR protein staining results, and this is proposed herein. CONCLUSION: There is wide support for universal LS screening in patients with EC, but this survey highlights areas of considerable variation in practice.


Assuntos
Neoplasias Colorretais Hereditárias sem Polipose/patologia , Reparo de Erro de Pareamento de DNA , Neoplasias do Endométrio/patologia , Neoplasias Ovarianas/patologia , Idoso , Neoplasias Colorretais Hereditárias sem Polipose/diagnóstico , Neoplasias do Endométrio/diagnóstico , Feminino , Humanos , Imuno-Histoquímica , Pessoa de Meia-Idade , Neoplasias Ovarianas/diagnóstico , Inquéritos e Questionários
10.
Shock ; 48(4): 401-410, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28915215

RESUMO

Clinical evidence reveals the existence of a trauma-induced secondary cardiac injury (TISCI) that is associated with poor patient outcomes. The mechanisms leading to TISCI in injured patients are uncertain. Conversely, animal models of trauma hemorrhage have repeatedly demonstrated significant cardiac dysfunction following injury, and highlighted mechanisms through which this might occur. The aim of this review was to provide an overview of the animal studies describing TISCI and its pathophysiology.Basic science models of trauma show evidence of innate immune system activation via Toll-like receptors, the exact protagonists of which remain unclear. Shortly following trauma and hemorrhage, cardiomyocytes upregulate gene regulatory protein and inflammatory molecule expression including nuclear factor kappa beta, tumor necrosis factor alpha, and interleukin-6. This is associated with expression of membrane bound adhesion molecules and chemokines leading to marked myocardial leukocyte infiltration. This cell activation and infiltration is linked to a rise in enzymes that cause oxidative and nitrative stress and subsequent protein misfolding within cardiomyocytes. Such protein damage may lead to reduced contractility and myocyte apoptosis. Other molecules have been identified as cardioprotective following injury. These include p38 mitogen-activated protein kinases and heat shock proteins.The balance between increasing damaging mediators and a reduction in cardio-protective molecules appears to define myocardial function following trauma. Exogenous therapeutics have been trialled in rodents with promising abilities to favorably alter this balance, and subsequently lead to improved cardiac function.


Assuntos
Traumatismos Cardíacos/metabolismo , Miocárdio/metabolismo , Choque Hemorrágico/metabolismo , Ferimentos e Lesões/metabolismo , Animais , Modelos Animais de Doenças , Traumatismos Cardíacos/etiologia , Traumatismos Cardíacos/patologia , Humanos , Miocárdio/patologia , Choque Hemorrágico/complicações , Choque Hemorrágico/patologia , Ferimentos e Lesões/complicações , Ferimentos e Lesões/patologia
11.
J Trauma Acute Care Surg ; 79(1): 71-7, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26091317

RESUMO

BACKGROUND: Secondary cardiac injury and dysfunction may be important contributors to poor outcomes in trauma patients, but the pathophysiology and clinical impact remain unclear. Early elevations in cardiac injury markers have been associated with the development of adverse cardiac events (ACEs), prolonged intensive care unit stays, and increased mortality. Studies of preinjury ß-blocker use suggest a potential protective effect in critically ill trauma patients. This study aimed to prospectively examine the association of early biomarker evidence of trauma-induced secondary cardiac injury (TISCI) and ACEs and to examine the potential contribution of circulating catecholamines to its pathophysiology. METHOD: Injured patients who met the study criteria were recruited at a single major trauma center. A blood sample was collected immediately on arrival. Serum epinephrine (E), norepinephrine (NE), and cardiac biomarkers including heart-related fatty acid binding protein (H-FABP) were assayed. Data were prospectively collected on ACEs. RESULTS: Of 300 patients recruited, 38 (13%) developed an ACE and had increased mortality (19% vs. 9%, p = 0.01) and longer intensive care unit stays (13 days, p < 0.001). H-FABP was elevated on admission in 56% of the patients, predicted the development of ACE, and was associated with higher mortality (14% vs. 5%, p = 0.01). Admission E and NE levels were strongly associated with elevations in H-FABP and ACEs (E, 274.0 pg/mL vs. 622.5 pg/mL, p < 0.001; NE, 1,063.2 pg/mL vs. 2,032.6 pg/mL, p < 0.001). Catecholamine effect on the development of TISCI or ACEs was not statistically independent of injury severity or depth of shock. CONCLUSION: Admission levels of H-FABP predict the development of ACEs and may be useful for prognosis and stratification of trauma patients. The development of TISCI and ACEs was associated with high admission levels of catecholamines, but their role in pathogenesis remains unclear. Clinical trials of adrenergic blockade may have the potential to demonstrate outcomes in patients presenting with TISCI. LEVEL OF EVIDENCE: Prognostic/epidemiologic study, level II.


Assuntos
Biomarcadores/sangue , Doenças Cardiovasculares/epidemiologia , Catecolaminas/sangue , Traumatismos Cardíacos/sangue , Traumatismos Cardíacos/complicações , Traumatismos Cardíacos/fisiopatologia , Ferimentos e Lesões/sangue , Ferimentos e Lesões/complicações , Adulto , Idoso , Idoso de 80 Anos ou mais , Traumatismos Cardíacos/epidemiologia , Humanos , Pessoa de Meia-Idade , Prognóstico , Estudos Prospectivos , Fatores de Risco , Ferimentos e Lesões/fisiopatologia , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA