Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Arch Pathol Lab Med ; 147(6): 643-654, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-36161544

RESUMEN

CONTEXT.­: Plasmablastic lymphoma (PBL) is a rare aggressive lymphoma, usually positive for CD138 and frequently occurring in the oral cavity of human immunodeficiency virus (HIV) patients. Up to 10% of cases are negative for CD138 and diagnostically very challenging. OBJECTIVE.­: To investigate the appropriate approach to diagnose CD138- plasmablastic lymphoma and avoid misdiagnosis. DESIGN.­: We studied 21 cases of CD138- PBL from multiple large institutes in the United States and 21 cases from the literature. RESULTS.­: CD138- PBLs were positive for different B/plasma cell markers at various percentages: MUM1 (94.4%; 34 of 36), OCT2 (70.6%; 12 of 17), immunoglobulin light chains (68.8%; 22 of 32), CD38 (68.4%; 13 of 19), CD79a (34.2%; 13 of 38), and PAX5 (15.6%; 5 of 32), suggesting that MUM1, OCT2, immunoglobulin light chains, and CD38 are useful markers to help establish the lineage. A total of 83% of cases (30 of 36) were extraoral lesions. Extraoral lesions showed much lower Epstein-Barr virus (EBV) infection rates (16 of 30; 53.3%) and had worse prognosis. MYC was positive in 80% (8 of 10) of EBV+ cases and 40% (2 of 5) EBV- cases, indicating the importance of MYC in pathogenesis, especially in EBV+ cases. CONCLUSIONS.­: Our study emphasizes that CD138- PBLs tend to be extraoral lesions, with much lower EBV infection rates, and diagnostically very challenging. Accurate diagnosis requires a thorough investigation and workup by using appropriate markers.


Asunto(s)
Infecciones por Virus de Epstein-Barr , Linfoma Inmunoblástico de Células Grandes , Linfoma Plasmablástico , Humanos , Linfoma Plasmablástico/diagnóstico , Linfoma Plasmablástico/patología , Infecciones por Virus de Epstein-Barr/complicaciones , Infecciones por Virus de Epstein-Barr/diagnóstico , Infecciones por Virus de Epstein-Barr/patología , Herpesvirus Humano 4 , Linfoma Inmunoblástico de Células Grandes/patología , Cadenas Ligeras de Inmunoglobulina , Estudios Multicéntricos como Asunto
2.
Eur J Pharmacol ; 891: 173780, 2021 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-33271152

RESUMEN

Paclitaxel is widely used in the treatment of breast, ovarian, lung, and other cancers. Its primary mechanism is to prevent microtubule depolymerization causing loss of dynamic instability crucial for normal microtubule function leading to mitotic arrest. Prolonged mitotic arrest results in cell death as a secondary response. The effects of paclitaxel are typically studied in cell lines which precludes assessment of the possible influence of tumor-associated cells. We therefore examined paclitaxel action ex vivo in fresh explant cultures of human breast tumors. Surprisingly, we found that paclitaxel failed to induce tumor cell death in explant culture, in contrast to several other cytotoxic agents including salinomycin and vincristine. The lack of effect was not due to defective drug uptake, and furthermore, analysis of H&E stained tumor slices indicated that paclitaxel treatment caused defective (granular) mitosis and chromosomal condensation in 5-10% of tumor cells after 72 h. These results suggest that while paclitaxel was able to penetrate into the tumor slice and disrupt mitosis in cycling tumor cells, any ensuing cell death likely occurred beyond the useful lifetime of the tumor slices. We conclude that explant culture systems may be inappropriate for the study of cytotoxic drugs where a delay exists between the drug's primary and secondary modes of action.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Mitosis/efectos de los fármacos , Paclitaxel/farmacología , Antineoplásicos/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Muerte Celular/efectos de los fármacos , Femenino , Humanos , Células MCF-7 , Paclitaxel/metabolismo , Piranos/farmacología , Factores de Tiempo , Técnicas de Cultivo de Tejidos , Vincristina/farmacología
3.
Antioxidants (Basel) ; 9(5)2020 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-32403251

RESUMEN

In patients with abdominal region cancers, ionizing radiation (IR)-induced long-term liver injury is a major limiting factor in the use of radiotherapy. Previously, the major mitochondrial deacetylase, sirtuin 3 (SIRT3), has been implicated to play an important role in the development of acute liver injury after total body irradiation but no studies to date have examined the role of SIRT3 in liver's chronic response to radiation. In the current study, ten-month-old Sirt3-/- and Sirt3+/+ male mice received 24 Gy radiation targeted to liver. Six months after exposure, irradiated Sirt3-/- mice livers demonstrated histopathological elevations in inflammatory infiltration, the loss of mature bile ducts and higher DNA damage (TUNEL) as well as protein oxidation (3-nitrotyrosine). In addition, increased expression of inflammatory chemokines (IL-6, IL-1ß, TGF-ß) and fibrotic factors (Procollagen 1, α-SMA) were also measured in Sirt3-/- mice following 24 Gy IR. The alterations measured in enzymatic activities of catalase, glutathione peroxidase, and glutathione reductase in the livers of irradiated Sirt3-/- mice also implied that hydrogen peroxide and hydroperoxide sensitive signaling cascades in the absence of SIRT3 might contribute to the IR-induced long-term liver injury.

4.
J Biochem Mol Toxicol ; 34(6): e22487, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32141170

RESUMEN

Colchicine (COL) shows strong anticancer activity but due to its toxicity towards normal cells its wider application is limited. To address this issue, a library of 17 novel COL derivatives, namely N-carbamates of N-deacetyl-4-(bromo/chloro/iodo)thiocolchicine, has been tested against two types of primary cancer cells. These included acute lymphoblastic leukemia (ALL) and human breast cancer (BC) derived from two different tumor subtypes, ER+ invasive ductal carcinoma grade III (IDCG3) and metastatic carcinoma (MC). Four novel COL derivatives showed higher anti-proliferative activity than COL (IC50 = 8.6 nM) towards primary ALL cells in cell viability assays (IC50 range of 1.1-6.4 nM), and several were more potent towards primary IDCG3 (IC50 range of 0.1 to 10.3 nM) or MC (IC50 range of 2.3-9.1 nM) compared to COL (IC50 of 11.1 and 11.7 nM, respectively). In addition, several derivatives were selectively active toward primary breast cancer cells compared to normal breast epithelial cells. The most promising derivatives were subsequently tested against the NCI panel of 60 human cancer cell lines and seven derivatives were more potent than COL against leukemia, non-small-cell lung, colon, CNS and prostate cancers. Finally, COL and two of the most active derivatives were shown to be effective in killing BC cells when tested ex vivo using fresh human breast tumor explants. The present findings indicate that the select COL derivatives constitute promising lead compounds targeting specific types of cancer.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Carbamatos/farmacología , Carcinoma Ductal de Mama/metabolismo , Colchicina/análogos & derivados , Extractos Vegetales/farmacología , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Apoptosis/efectos de los fármacos , Carcinoma Ductal de Mama/patología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Colchicina/farmacología , Colchicum/química , Ensayos de Selección de Medicamentos Antitumorales/métodos , Femenino , Humanos , Concentración 50 Inhibidora , Células MCF-7 , Persona de Mediana Edad , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Receptores de Estrógenos/metabolismo , Neoplasias de la Mama Triple Negativas/patología
5.
Blood ; 135(2): 108-120, 2020 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-31697816

RESUMEN

NF-κB and Notch signaling can be simultaneously activated in a variety of B-cell lymphomas. Patients with B-cell lymphoma occasionally develop clonally related myeloid tumors with poor prognosis. Whether concurrent activation of both pathways is sufficient to induce B-cell transformation and whether the signaling initiates B-myeloid conversion in a pathological context are largely unknown. Here, we provide genetic evidence that concurrent activation of NF-κB and Notch signaling in committed B cells is sufficient to induce B-cell lymphomatous transformation and primes common progenitor cells to convert to myeloid lineage through dedifferentiation, not transdifferentiation. Intriguingly, the converted myeloid cells can further transform, albeit at low frequency, into myeloid leukemia. Mechanistically, coactivation of NF-κB and Notch signaling endows committed B cells with the ability to self renew. Downregulation of BACH2, a lymphoma and myeloid gene suppressor, but not upregulation of CEBPα and/or downregulation of B-cell transcription factors, is an early event in both B-cell transformation and myeloid conversion. Interestingly, a DNA hypomethylating drug not only effectively eliminated the converted myeloid leukemia cells, but also restored the expression of green fluorescent protein, which had been lost in converted myeloid leukemia cells. Collectively, our results suggest that targeting NF-κB and Notch signaling will not only improve lymphoma treatment, but also prevent the lymphoma-to-myeloid tumor conversion. Importantly, DNA hypomethylating drugs might efficiently treat these converted myeloid neoplasms.


Asunto(s)
Linfocitos B/patología , Transformación Celular Neoplásica/patología , Linfoma de Células B de la Zona Marginal/patología , Células Mieloides/patología , FN-kappa B/metabolismo , Receptores Notch/metabolismo , Animales , Linfocitos B/metabolismo , Transformación Celular Neoplásica/metabolismo , Femenino , Humanos , Linfoma de Células B de la Zona Marginal/genética , Linfoma de Células B de la Zona Marginal/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Células Mieloides/metabolismo , FN-kappa B/genética , Receptores Notch/genética , Transducción de Señal
6.
PLoS One ; 14(11): e0224621, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31714922

RESUMEN

Classic Hodgkin lymphoma (CHL) characteristically shows few malignant cells in a microenvironment comprised of mixed inflammatory cells. Although CHL is associated with a high cure rate, recent studies have associated poor prognosis with absolute monocyte count in peripheral blood and increased monocyte/macrophages in involved lymph nodes. Thus, the role of monocytic infiltration and macrophage differentiation in the tumor microenvironment of CHL may be more relevant than absolute macrophage numbers to defining prognosis in CHL patients and potentially have therapeutic implications. Most studies identify tumor-associated macrophages (TAMs) using markers (e.g., CD68) expressed by macrophages and other mononuclear phagocytes, such as monocytes. In contrast, Class A Scavenger Receptor (SR-A/CD204) is expressed by tissue macrophages but not monocytic precursors. In this study, we examined SR-A expression in CHL (n = 43), and compared its expression with that of other macrophage markers. We confirmed a high prevalence of mononuclear cells that stained with CD68, CD163, and CD14 in CHL lymph nodes. However, SR-A protein expression determined by immunohistochemistry was limited to macrophages localized in sclerotic bands characteristic of nodular sclerosis CHL. In contrast, SR-A protein was readily detectable in lymph nodes with metastatic tumor, extra-nodal CHL, T cell/histiocyte-rich large B cell lymphoma, and resident macrophages in non-malignant tissues, including spleen, lymph node, liver and lung. The results of SR-A protein expression paralleled the expression of SR-A mRNA determined by quantitative RT-PCR. These data provide evidence that tumor-infiltrating monocyte/macrophages in CHL have a unique phenotype that likely depends on the microenvironment of nodal CHL.


Asunto(s)
Antígenos CD/metabolismo , Enfermedad de Hodgkin/patología , Monocitos/patología , Microambiente Tumoral/fisiología , Enfermedad de Hodgkin/metabolismo , Humanos , Macrófagos/metabolismo , Macrófagos/patología , Monocitos/metabolismo , Fenotipo , Estudios Retrospectivos
7.
Oncol Lett ; 18(5): 5097-5106, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31612021

RESUMEN

Phyllodes tumors of the breast (PTB) are uncommon stromal-epithelial neoplasms, with the main recommended treatment being surgical removal. However, even with adequate resection, the risk of recurrence in the malignant form remains as high as 40%, and there is no recognized consensus on the most effective drugs for PTB. In the present study, an ex vivo model of malignant phyllodes and derived primary cell cultures were used to evaluate the effectiveness of a panel of different drugs, including the Bcl-2/Bcl-xL inhibitor ABT-263, salinomycin (SAL), doxorubicin (DOX), paclitaxel (TAX), vincristine (VCR), colchicine (COL) and cisplatin (CIS). ABT-263, SAL and DOX were highly effective towards phyllodes spindle cells when assessed in the ex vivo model, contributing to ~98% tumor cell death. Furthermore, ABT-263 was highly selective for tumor cells in this system, and exhibited little toxic effect on adjacent normal epithelial cells. Furthermore, consistent with findings in the ex vivo model, ABT-263 was significantly less toxic towards MCF 10A non-tumorigenic breast epithelial cells compared with SAL and DOX. A conditional reprogramming strategy was subsequently used, involving Rho kinase inhibition, to successfully generate primary phyllodes tumor cells that could be cultured for several passages. The primary cells were sensitive to DOX with an IC50 of 0.40±0.07 µM in a standard viability assay and the preliminary results were obtained indicating sensitivity to ABT-263 and SAL. The present study demonstrated the feasibility of using explants and primary cells for drug discovery, selectively targeting PTB cells.

8.
Nature ; 572(7768): 260-264, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31341286

RESUMEN

In humans, the adaptive immune system uses the exchange of information between cells to detect and eliminate foreign or damaged cells; however, the removal of unwanted cells does not always require an adaptive immune system1,2. For example, cell selection in Drosophila uses a cell selection mechanism based on 'fitness fingerprints', which allow it to delay ageing3, prevent developmental malformations3,4 and replace old tissues during regeneration5. At the molecular level, these fitness fingerprints consist of combinations of Flower membrane proteins3,4,6. Proteins that indicate reduced fitness are called Flower-Lose, because they are expressed in cells marked to be eliminated6. However, the presence of Flower-Lose isoforms at a cell's membrane does not always lead to elimination, because if neighbouring cells have similar levels of Lose proteins, the cell will not be killed4,6,7. Humans could benefit from the capability to recognize unfit cells, because accumulation of damaged but viable cells during development and ageing causes organ dysfunction and disease8-17. However, in Drosophila this mechanism is hijacked by premalignant cells to gain a competitive growth advantage18. This would be undesirable for humans because it might make tumours more aggressive19-21. It is unknown whether a similar mechanism of cell-fitness comparison is present in humans. Here we show that two human Flower isoforms (hFWE1 and hFWE3) behave as Flower-Lose proteins, whereas the other two isoforms (hFWE2 and hFWE4) behave as Flower-Win proteins. The latter give cells a competitive advantage over cells expressing Lose isoforms, but Lose-expressing cells are not eliminated if their neighbours express similar levels of Lose isoforms; these proteins therefore act as fitness fingerprints. Moreover, human cancer cells show increased Win isoform expression and proliferate in the presence of Lose-expressing stroma, which confers a competitive growth advantage on the cancer cells. Inhibition of the expression of Flower proteins reduces tumour growth and metastasis, and induces sensitivity to chemotherapy. Our results show that ancient mechanisms of cell recognition and selection are active in humans and affect oncogenic growth.


Asunto(s)
Canales de Calcio/metabolismo , Proliferación Celular , Proteínas de Drosophila/metabolismo , Neoplasias/patología , Isoformas de Proteínas/metabolismo , Animales , Canales de Calcio/genética , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Drosophila melanogaster , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Metástasis de la Neoplasia , Neoplasias/tratamiento farmacológico , Isoformas de Proteínas/genética
9.
PLoS Negl Trop Dis ; 12(10): e0006727, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30286207

RESUMEN

A young, healthy traveler returning to the United States presented with fever, night sweats, splenomegaly, and pancytopenia. Bone marrow biopsy revealed leishmaniasis (Leishmania infantum), likely acquired in southern France. Although many cases of endemic visceral leishmaniasis (VL) have been reported in Europe, this is a rare case of imported VL in a healthy traveler returning from Europe to the US. Despite successful initial treatment with liposomal amphotericin B (LamB), relapse occurred. Treatments for VL in immunocompetent individuals are highly effective, but relapse can occur. There is more extensive experience in endemic areas with treating relapse that may be lacking in North America. This case alerts physicians in the US that immunocompetent adults can acquire VL during brief visits to endemic areas in Europe. It is important that travelers be counseled on preventive measures. Patients should be monitored after treatment for relapse.


Asunto(s)
Enfermedades Transmisibles Importadas/diagnóstico , Enfermedades Transmisibles Importadas/patología , Leishmania infantum/aislamiento & purificación , Leishmaniasis Visceral/diagnóstico , Leishmaniasis Visceral/patología , Viaje , Anfotericina B/uso terapéutico , Antiprotozoarios/uso terapéutico , Biopsia , Médula Ósea/parasitología , Enfermedades Transmisibles Importadas/tratamiento farmacológico , Enfermedades Transmisibles Importadas/parasitología , Francia , Humanos , Leishmaniasis Visceral/tratamiento farmacológico , Leishmaniasis Visceral/parasitología , Masculino , Recurrencia , Estados Unidos , Adulto Joven
10.
Eur J Med Chem ; 156: 510-523, 2018 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-30025346

RESUMEN

The polyether ionophore salinomycin has recently captured much interest due to its potent activity against multi-drug resistant cancer cells and cancer stem cells. Previous studies have shown that either acylation of the C20 position or esterification/amidation of the C1 carboxylate moiety is beneficial in terms of biological properties. In this paper, we present the first analogs combining such modifications. Evaluation of the anti-proliferative activity against a series of cancer cell lines showed that acylation of the C20 hydroxyl group improves the activity of salinomycin C1 amides but not of the corresponding C1 esters. Importantly, the activity of several of the doubly modified analogs surpasses that of commonly used cytostatic drugs cisplatin and doxorubicin in the LoVo/DX multi-drug resistant cell line. All analogs were tested against primary acute lymphoblastic leukemia cells in standard cell viability assays; three were more potent than salinomycin. Further studies revealed that selected analogs induced characteristics of apoptotic cell death and increased expression of p53. Additionally, using an ex vivo model of breast tumor, tumor cell viability significantly decreased after treatment with salinomycin or its double-modified derivative (3a) in a time-dependent manner. The present findings indicate that double-modified salinomycin derivatives constitute promising lead compounds for targeting various types of cancer.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Piranos/química , Piranos/farmacología , Anciano , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Células Tumorales Cultivadas
11.
Diagn Pathol ; 13(1): 15, 2018 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-29463311

RESUMEN

BACKGROUND: Plasma cell myeloma (PCM) is a neoplasm of terminally differentiated B lymphocytes with molecular heterogeneity. Although therapy-related myeloid neoplasms are common in plasma cell myeloma patients after chemotherapy, transdifferentiation of plasma cell myeloma into myeloid neoplasms has not been reported in literature. Here we report a very rare case of myeloid neoplasm transformed from plasma cell myeloma. CASE PRESENTATION: A 60-year-old man with a history of plasma cell myeloma with IGH-MAF gene rearrangement and RAS/RAF mutations developed multiple soft tissue lesions one year following melphalan-based chemotherapy and autologous stem cell transplant. Morphological and immunohistochemical characterization of the extramedullary disease demonstrated that the tumor cells were derived from the monocyte-macrophage lineage. Next generation sequencing (NGS) studies detected similar clonal aberrations in the diagnostic plasma cell population and post-therapy neoplastic cells, including IGH-MAF rearrangement, multiple genetic mutations in RAS signaling pathway proteins, and loss of tumor suppressor genes. Molecular genetic analysis also revealed unique genomic alterations in the transformed tumor cells, including gain of NF1 and loss of TRAF3. CONCLUSION: To our knowledge, this is the first case of myeloid sarcoma transdifferentiated from plasma cell neoplasm. Our findings in this unique case suggest clonal evolution of plasma cell myeloma to myeloma neoplasm and the potential roles of abnormal RAS/RAF signaling pathway in lineage switch or transdifferentiation.


Asunto(s)
Evolución Clonal/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Mieloma Múltiple/genética , Células Plasmáticas/patología , Transformación Celular Neoplásica , Aberraciones Cromosómicas , Humanos , Masculino , Persona de Mediana Edad , Mieloma Múltiple/diagnóstico , Mieloma Múltiple/patología , Patología Molecular/métodos
12.
Mod Pathol ; 31(5): 718-731, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29327711

RESUMEN

Rare B-cell neoplasms with plasmablastic differentiation may aberrantly express CD3 by immunohistochemical staining, which places a great challenge for diagnosis. We here studied 17 cases of CD3+ plasmablastic B-cell neoplasms, including 12 plasmablastic lymphomas and 5 plasmablastic plasma cell myelomas. All 17 cases occurred in the extranodal sites with a male predominance (13/17). Four cases were initially misinterpreted by outside institutions, among which three were diagnosed as 'peripheral T-cell lymphoma, not otherwise specified' and one was classified as 'poorly differentiated neuroendocrine carcinoma'. The plasmablastic cells were present in all 17 cases diffusely or in a subset of tumor cells. CD3 expression was mostly diffuse (12/17) and moderate to strong (11/16) with a cytoplasmic staining pattern (14/16). Other T-cell markers were nearly absent, including CD2 (0/10), CD4 (1/13), CD5 (0/14), CD7 (0/11), and CD8 (0/13). CD138 was positive in all 17 cases and CD79a was variably positive in 8 of 14 cases. Only one case had immunoreactivity to CD20 (1/17) and PAX5 (1/12). CD56 expression and EBV infection were detected in 8/15 and 6/17, respectively. No HHV8 infection was noted in all 11 cases tested. Most cases (11/13) revealed either kappa or lambda light chain restriction. Of the nine cases studied, six had clonal IGH rearrangements but no clonal TRG rearrangements. Our study further emphasizes that the accurate classification of CD3+ plasmablastic neoplasms requires thorough morphologic examination, incorporation of more B-cell and T-cell markers in addition to CD3 and CD20, frequent addition of CD138 staining, and utilization of necessary molecular and genetic studies.


Asunto(s)
Complejo CD3 , Linfoma de Células B/diagnóstico , Mieloma Múltiple/diagnóstico , Linfoma Plasmablástico/diagnóstico , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Linfocitos B/patología , Biomarcadores de Tumor , Diagnóstico Diferencial , Femenino , Humanos , Linfoma de Células B/patología , Masculino , Persona de Mediana Edad , Mieloma Múltiple/patología , Linfoma Plasmablástico/patología , Factores Sexuales , Adulto Joven
13.
Ann Clin Lab Sci ; 46(5): 474-9, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27650613

RESUMEN

The objective of the study was to evaluate the expression pattern of interleukin-6 (IL-6) to determine its utility in differentiating Castleman Disease subtypes and reactive lymphadenopathies. Paraffin-embedded tissue blocks from 20 cases referred for assessment of Castleman Disease (CD) and 4 cases of reactive hyperplasia were selected for immunohistochemical staining with an IL-6 antibody. Six pathologists evaluated the hematoxylin and eosin stained tissue sections and IL-6 expression pattern. Of 20 CD referral cases, the pathologic diagnosis was CD in 14 cases and included 6 hyaline-vascular (HV-CD), 6 plasma cell (PC-CD) and 2 "mixed type"-CD cases. The remaining 6 referral cases showed morphologic features consistent with reactive lymphadenopathy. Patients with non-CD, reactive lymphadenopathies had clinical and/or laboratory features of systemic lupus erythematosus, Hashimoto's disease, viral infection or chronic cellulitis. The pattern of IL-6 expression differed between CD subtypes and non-CD cases. In PC-CD, IL-6 expression was detected in plasma cells and vascular endothelial cells; whereas IL-6 immunoreactivity was detected primarily in vascular endothelial cells in HV-CD. Interfollicular plasma cells were prominent in PC-CD and reactive lymphadenopathies; however, IL-6 expression was significantly increased in PC-CD compared to reactive lymph nodes. Together with morphologic features, the expression pattern of IL-6 detected by immunohistochemistry is helpful to distinguish CD subtypes and reactive mimics.


Asunto(s)
Enfermedad de Castleman/clasificación , Enfermedad de Castleman/diagnóstico , Interleucina-6/metabolismo , Linfadenopatía/diagnóstico , Biopsia , Femenino , Humanos , Inmunohistoquímica , Ganglios Linfáticos/patología , Masculino , Persona de Mediana Edad , Células Plasmáticas/metabolismo
14.
Nat Commun ; 6: 6328, 2015 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-25692908

RESUMEN

Among cyclin-dependent kinase inhibitors that control the G1 phase in cell cycle, only p18 and p27 can negatively regulate haematopoietic stem cell (HSC) self-renewal. In this manuscript, we demonstrate that p18 protein is a more potent inhibitor of HSC self-renewal than p27 in mouse models and its deficiency promoted HSC expansion in long-term culture. Single-cell analysis indicated that deleting p18 gene favoured self-renewing division of HSC in vitro. Based on the structure of p18 protein and in-silico screening, we further identified novel smallmolecule inhibitors that can specifically block the activity of p18 protein. Our selected lead compounds were able to expand functional HSCs in a short-term culture. Thus, these putative small-molecule inhibitors for p18 protein are valuable for further dissecting the signalling pathways of stem cell self-renewal and may help develop more effective chemical agents for therapeutic expansion of HSC.


Asunto(s)
Técnicas de Cultivo de Célula , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/metabolismo , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/citología , Animales , Apoptosis , Ciclo Celular , Proliferación Celular , Separación Celular , Técnicas de Cocultivo , Quinasa 6 Dependiente de la Ciclina/metabolismo , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/antagonistas & inhibidores , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Femenino , Eliminación de Gen , Células Madre Hematopoyéticas/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/metabolismo , Fenotipo , Transducción de Señal , Análisis de la Célula Individual , Células Madre/citología
15.
Nat Commun ; 4: 2174, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23873265

RESUMEN

Mechanisms underlying the reprogramming process of induced pluripotent stem cells remain poorly defined. Like tumorigenesis, generation of induced pluripotent stem cells was shown to be suppressed by the Trp53 (p53) pathway, at least in part via p21Cdkn1a (p21)-mediated cell cycle arrest. Here we examine the role of PUMA, a pro-apoptotic mediator of p53, during somatic reprogramming in comparison to p21 in the p53 pathway. Using mouse strains deficient in these molecules, we demonstrate that PUMA is an independent mediator of the negative effect of p53 on induced pluripotent stem cell induction. PUMA deficiency leads to a better survival rate associated with reduced DNA damage and fewer chromosomal aberrations in induced pluripotent stem cells, whereas loss of p21 or p53 results in an opposite outcome. Given these new findings, PUMA may serve as a distinct and more desirable target in the p53 pathway for induced pluripotent stem cell generation, thereby having important implications for potential therapeutic applications of induced pluripotent stem cells.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células Madre Pluripotentes Inducidas/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteínas Supresoras de Tumor/genética , Animales , Proteínas Reguladoras de la Apoptosis/deficiencia , Biomarcadores/metabolismo , Ciclo Celular/genética , Diferenciación Celular , Linaje de la Célula/genética , Aberraciones Cromosómicas , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/deficiencia , Daño del ADN , Embrión de Mamíferos , Femenino , Fibroblastos/citología , Eliminación de Gen , Células Madre Pluripotentes Inducidas/citología , Masculino , Ratones , Ratones Transgénicos , Transducción de Señal , Proteína p53 Supresora de Tumor/deficiencia , Proteínas Supresoras de Tumor/deficiencia
16.
Blood ; 119(15): 3629-37, 2012 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-22374698

RESUMEN

Ultimate success of hematopoietic stem cell transplantation (HSCT) depends not only on donor HSCs themselves but also on the host environment. Total body irradiation is a component in various host conditioning regimens for HSCT. It is known that ionizing radiation exerts "bystander effects" on nontargeted cells and that HSCs transplanted into irradiated recipients undergo proliferative exhaustion. However, whether irradiated recipients pose a proliferation-independent bystander effect on transplanted HSCs is unclear. In this study, we found that irradiated mouse recipients significantly impaired the long-term repopulating ability of transplanted mouse HSCs shortly (∼ 17 hours) after exposure to irradiated hosts and before the cells began to divide. There was an increase of acute cell death associated with accelerated proliferation of the bystander hematopoietic cells. This effect was marked by dramatic down-regulation of c-Kit, apparently because of elevated reactive oxygen species. Administration of an antioxidant chemical, N-acetylcysteine, or ectopically overexpressing a reactive oxygen species scavenging enzyme, catalase, improved the function of transplanted HSCs in irradiated hosts. Together, this study provides evidence for an acute negative, yet proliferation-independent, bystander effect of irradiated recipients on transplanted HSCs, thereby having implications for HSCT in both experimental and clinical scenarios in which total body irradiation is involved.


Asunto(s)
Efecto Espectador/efectos de la radiación , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/efectos de la radiación , Traumatismos Experimentales por Radiación/etiología , Enfermedad Aguda , Animales , Efecto Espectador/inmunología , Células Cultivadas , Células HEK293 , Células Madre Hematopoyéticas/fisiología , Humanos , Ratones , Ratones Congénicos , Ratones Endogámicos C57BL , Modelos Biológicos , Traumatismos Experimentales por Radiación/inmunología , Acondicionamiento Pretrasplante/efectos adversos , Irradiación Corporal Total/efectos adversos
17.
Shock ; 37(5): 518-23, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22293600

RESUMEN

The release of hematopoietic progenitor cells (HPCs) from bone marrow (BM) is under tight homeostatic control. Under stress conditions, HPCs migrate from BM and egress into circulation to participate in immune response, wound repair, or tissue regeneration. Hemorrhagic shock with resuscitation (HS/R), resulting from severe trauma and major surgery, promotes HPC mobilization from BM, which, in turn, affects post-HS immune responses. In this study, we investigated the mechanism of HS/R regulation of HPC mobilization from BM. Using a mouse HS/R model, we demonstrate that the endogenous alarmin molecule high-mobility group box 1 mediates HS/R-induced granulocyte colony-stimulating factor secretion from macrophages (Mϕ in a RAGE [receptor for advanced glycation end products] signaling-dependent manner. Secreted granulocyte colony-stimulating factor, in turn, induces HPC egress from BM. We also show that activation of ß-adrenergic receptors on Mϕ by catecholamine mediates the HS/R-induced release of high-mobility group box 1. These data indicate that HS/R, a global ischemia-reperfusion stimulus, regulates HPC mobilization through a series of interacting pathways that include neuroendocrine and innate immune systems, in which Mϕ play a central role.


Asunto(s)
Médula Ósea/inmunología , Movimiento Celular/inmunología , Células Madre Hematopoyéticas/inmunología , Inmunidad Innata , Macrófagos/inmunología , Choque Hemorrágico/inmunología , Transducción de Señal/inmunología , Animales , Médula Ósea/patología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Proteína HMGB1/inmunología , Células Madre Hematopoyéticas/patología , Macrófagos/patología , Masculino , Ratones , Ratones Noqueados , Choque Hemorrágico/patología
18.
Blood ; 115(17): 3472-80, 2010 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-20177048

RESUMEN

Molecular paradigms underlying the death of hematopoietic stem cells (HSCs) induced by ionizing radiation are poorly defined. We have examined the role of Puma (p53 up-regulated mediator of apoptosis) in apoptosis of HSCs after radiation injury. In the absence of Puma, HSCs were highly resistant to gamma-radiation in a cell autonomous manner. As a result, Puma-null mice or the wild-type mice reconstituted with Puma-null bone marrow cells were strikingly able to survive for a long term after high-dose gamma-radiation that normally would pose 100% lethality on wild-type animals. Interestingly, there was no increase of malignancy in the exposed animals. Such profound beneficial effects of Puma deficiency were likely associated with better maintained quiescence and more efficient DNA repair in the stem cells. This study demonstrates that Puma is a unique mediator in radiation-induced death of HSCs. Puma may be a potential target for developing an effective treatment aimed to protect HSCs from lethal radiation.


Asunto(s)
Proteínas Reguladoras de la Apoptosis , Apoptosis/genética , Apoptosis/efectos de la radiación , Reparación del ADN/genética , Reparación del ADN/efectos de la radiación , Rayos gamma/efectos adversos , Células Madre Hematopoyéticas/metabolismo , Proteínas Supresoras de Tumor , Animales , Supervivencia Celular/genética , Supervivencia Celular/efectos de la radiación , Eliminación de Gen , Ratones , Ratones Transgénicos
19.
Am J Physiol Gastrointest Liver Physiol ; 297(2): G249-58, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19556359

RESUMEN

Hepatic preconditioning has emerged as a promising strategy of activating natural pathways to augment tolerance to liver ischemia-reperfusion (IR) injury. Liver-resident natural killer T (NKT) cells play an important role in modulating the local immune and inflammatory responses. This work was aimed to investigate whether preactivation of NKT cells could provide a beneficial "preconditioning" effect to ameliorate the subsequent hepatic IR injury. To selectively activate NKT cells, C57BL/6 mice were treated intraperitoneally with the glycolipid antigen alpha-galactosylceramide (alpha-GalCer) 1 h prior to hepatic ischemia. Significantly reduced liver IR injury was observed in mice pretreated with alpha- GalCer, and this protective effect was specifically abrogated by a CD1d blocking antibody. Serum TNF-alpha, IFN-gamma, and IL-13 levels were markedly increased shortly after alpha-GalCer injection. Pretreatment with a neutralizing antibody against TNF-alpha or IFN-gamma did not influence the protective effect of alpha-GalCer preconditioning, whereas preadministration of an IL-13 neutralizing antibody completely abolished the effect. Treatment with alpha-GalCer also led to an increased expression of adenosine A2A receptor (A2AR) in the liver, and blockade of A2AR by SH58261 diminished alpha-GalCer pretreatment-mediated attenuation of liver IR injury. In contrast, administration of the selective A2AR agonist CGS21680 reversed the counteracting effect of the IL-13 neutralizing antibody on alpha-GalCer preconditioning. Additionally, alpha-GalCer pretreatment was associated with a decreased neutrophil accumulation in the ischemic liver. These findings provide the first evidence that hepatic preconditioning by preactivation of NKT cells with alpha-GalCer protects the liver from IR injury via an IL-13 and adenosine A2AR-dependent mechanism.


Asunto(s)
Galactosilceramidas/farmacología , Interleucina-13/sangre , Isquemia/tratamiento farmacológico , Hígado/efectos de los fármacos , Activación de Linfocitos/efectos de los fármacos , Células T Asesinas Naturales/efectos de los fármacos , Receptor de Adenosina A2A/efectos de los fármacos , Daño por Reperfusión/prevención & control , Animales , Modelos Animales de Enfermedad , Galactosilceramidas/administración & dosificación , Inyecciones Intraperitoneales , Interferón gamma/sangre , Isquemia/inmunología , Isquemia/metabolismo , Isquemia/patología , Hígado/irrigación sanguínea , Hígado/inmunología , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Células T Asesinas Naturales/inmunología , Células T Asesinas Naturales/metabolismo , Infiltración Neutrófila/efectos de los fármacos , Receptor de Adenosina A2A/metabolismo , Daño por Reperfusión/inmunología , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Factor de Necrosis Tumoral alfa/sangre
20.
J Immunol ; 182(1): 572-80, 2009 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19109190

RESUMEN

Polymorphonuclear neutrophils (PMN) are critical innate immune effector cells that either protect the host or exacerbate organ dysfunction by migrating to injured or inflamed tissues. Resuscitated hemorrhagic shock following major trauma promotes the development of organ inflammation by priming PMN migration and activation in response to a second, often trivial, stimulus (a so-called "two hit" phenomenon). PMN mobilization from bone marrow supports a sustained, hemorrhagic shock/resuscitation (HS/R)-primed migration of PMN. We addressed the role and mechanism of HS/R in regulating PMN egress from bone marrow. We demonstrate that HS/R through the alarmin HMGB1 induces IL-23 secretion from macrophages in an autocrine and TLR4 signaling-dependent manner. In turn IL-23, through an IL-17 G-CSF-mediated mechanism, induces PMN egress from bone marrow. We also show that beta-adrenergic receptor activation by catecholamine of macrophages mediates the HS/R-induced release of HMGB1. These data indicate that HS/R, a global ischemia/reperfusion stimulus, regulates PMN mobilization through a series of interacting pathways that include neuroendocrine and innate and acquired immune systems. Blocking this novel signaling axis may present a novel therapeutic target for posttrauma inflammation.


Asunto(s)
Células de la Médula Ósea/inmunología , Inmunidad Innata , Sistemas Neurosecretores/inmunología , Infiltración Neutrófila/inmunología , Choque Hemorrágico/inmunología , Transducción de Señal/inmunología , Animales , Comunicación Autocrina/inmunología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Movimiento Celular/inmunología , Epinefrina/fisiología , Proteína HMGB1/metabolismo , Proteína HMGB1/fisiología , Interleucina-23/metabolismo , Interleucina-23/fisiología , Macrófagos Alveolares/metabolismo , Masculino , Ratones , Ratones Endogámicos C3H , Receptores Adrenérgicos beta/fisiología , Resucitación/métodos , Choque Hemorrágico/metabolismo , Choque Hemorrágico/patología , Receptor Toll-Like 4/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA