Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Invest New Drugs ; 37(2): 364-374, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30073464

RESUMEN

Despite the significant progress in the field of cancer therapeutics, the incidence of pancreatic cancer (PC) has continuously increased. One possible mechanism for this increasing burden is impaired drug delivery and drug resistance resulting from a unique tumor microenvironment and genetic mutations. Apratoxins are potent anticancer agents and cotranslational translocation inhibitors with potential therapeutic applications to treat cancers with active secretory pathways. Here, we developed apratoxin S10 (Apra S10) as an anti-pancreatic cancer agent which potently inhibited the growth of both established and patient-derived primary pancreatic cancer cells. We validated its mechanism of action on pancreatic cancer cells by demonstrating the downregulation of multiple receptor tyrosine kinases and inhibition of growth factor and cytokine secretion. Apra S10 also inhibited a number of cytokines secreted by stromal cells, suggesting that Apra S10 not only inhibited pancreatic cancer cell secretion, but also reduced the level of factors secreted by other cell types active within the tumor microenvironment. As Apra S10 tissue distribution indicated its high enrichment in pancreas tissue, an orthotopic pancreatic patient-derived xenograft mouse model that closely mimics the human pancreatic tumor microenvironment was for the first time used in apratoxin studies. Apra S10 showed promising antitumor effect in this pancreatic cancer model and this effect was mediated through anti-proliferation properties.


Asunto(s)
Adenocarcinoma/patología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Depsipéptidos/farmacología , Neoplasias Pancreáticas/patología , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/metabolismo , Animales , Femenino , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Int J Mol Sci ; 19(12)2018 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-30513792

RESUMEN

Cancer cachexia is a debilitating condition seen frequently in patients with pancreatic ductal adenocarcinoma (PDAC). The underlying mechanisms driving cancer cachexia are not fully understood but are related, at least in part, to the immune response to the tumor both locally and systemically. We hypothesize that there are unique differences in cytokine levels in the tumor microenvironment and systemic circulation between PDAC tumors and that these varying profiles affect the degree of cancer cachexia observed. Patient demographics, operative factors, oncologic factors, and perioperative data were collected for the two patients in the patient derived xenograft (PDX) model. Human pancreatic cancer PDX were created by implanting fresh surgical pancreatic cancer tissues directly into immunodeficient mice. At PDX end point, mouse tumor, spleen and muscle tissues were collected and weighed, muscle atrophy related gene expression measured, and tumor and splenic soluble proteins were analyzed. PDX models were created from surgically resected patients who presented with different degrees of cachexia. Tumor free body weight and triceps surae weight differed significantly between the PDX models and control (P < 0.05). Both PDX groups had increased atrophy related gene expression in muscle compared to control (FoxO1, Socs3, STAT3, Acvr2b, Atrogin-1, MuRF1; P < 0.05). Significant differences were noted in splenic soluble protein concentrations in 14 of 15 detected proteins in tumor bearing mice when compared to controls. Eight splenic soluble proteins were significantly different between PDX groups (P < 0.05). Tumor soluble proteins were significantly different between the two PDX groups in 15 of 24 detected proteins (P < 0.05). PDX models preserve the cachectic heterogeneity found in patients and are associated with unique cytokine profiles in both the spleen and tumor between different PDX. These data support the use of PDX as a strategy to study soluble cachexia protein markers and also further efforts to elucidate which cytokines are most related to cachexia in order to provide potential targets for immunotherapy.


Asunto(s)
Adenocarcinoma/complicaciones , Adenocarcinoma/metabolismo , Caquexia/complicaciones , Caquexia/metabolismo , Carcinoma Ductal Pancreático/complicaciones , Carcinoma Ductal Pancreático/metabolismo , Citocinas/metabolismo , Medicina de Precisión , Adenocarcinoma/genética , Adenocarcinoma/patología , Anciano , Animales , Atrofia , Peso Corporal , Caquexia/genética , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Persona de Mediana Edad , Proteínas de Neoplasias/metabolismo , Solubilidad , Bazo/patología , Microambiente Tumoral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
3.
PLoS One ; 18(2): e0281182, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36812168

RESUMEN

In pancreatic cancer clinical trials, Black patients are under-represented while having higher morbidity and mortality rates as compared to other racial groups. Multiple factors, including socioeconomic and lifestyle factors may contribute to this disparity, but genomic contributions remain unclear. In an exploratory project to identify genes that may contribute to differences in survival between Black (n = 8) and White (n = 20) patients with pancreatic cancer, transcriptomic sequencing of over 24,900 genes was performed in human pancreatic tumor and non-tumor tissue obtained from Black and White patients. Over 4,400 genes were differentially expressed in tumor and non-tumor tissue, irrespective of race. To validate these results, the expression of four genes (AGR2, POSTN, TFF1, and CP) reported to be up-regulated in pancreatic tumor tissue as compared to non-tumor tissue were confirmed using quantitative PCR. Transcriptomic analysis that compared pancreatic tumor tissue from Black and White patients revealed differential expression in 1,200 genes, while a comparison of the non-tumor and tumor gene expression differences within each race revealed over 1,500 tumor-specific differentially expressed genes in pancreatic tumor and non-tumor tissue from Black patients. We identified TSPAN8 as a potential tumor-specific gene significantly overexpressed in pancreatic tumor tissue in Black patients as compared to White patients. Using Ingenuity Pathway Analysis software to compare the race-associated gene expression profiles, over 40 canonical pathways were identified to be potentially impacted by the gene expression differences between the races. Heightened expression of TSPAN8 was associated with poor overall survival, suggesting TSPAN8 as one potential genetic factor contributing to the differential outcomes in Black patients with pancreatic cancer, supporting the potential utility of larger genomic studies to further explore the role of TSPAN8 in pancreatic cancer.


Asunto(s)
Adenocarcinoma , Neoplasias Pancreáticas , Humanos , Mucoproteínas/genética , Proteínas Oncogénicas/genética , Neoplasias Pancreáticas/patología , Tetraspaninas/genética , Transcriptoma , Población Blanca , Población Negra , Neoplasias Pancreáticas
4.
Surg Oncol Clin N Am ; 31(4): 609-629, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36243497

RESUMEN

During an esophagectomy, many factors influence the anastomosis. Surgical factors include anastomotic tension, location of the anastomosis, surgical technique, and perfusion of the conduit. The use of fluorescent angiography is a possible avenue for more objective evaluation of the gastric conduit. There is a lot of variability in the way this tool has been used and what the results indicate. This article will discuss the various methods of fluorescent angiography to determine intestinal perfusion using indocyanine green and fluorescent imaging and the data on the association with clinical outcomes.


Asunto(s)
Neoplasias Esofágicas , Esofagectomía , Anastomosis Quirúrgica/métodos , Fuga Anastomótica/prevención & control , Fuga Anastomótica/cirugía , Neoplasias Esofágicas/cirugía , Esofagectomía/métodos , Humanos , Verde de Indocianina , Estómago/cirugía
5.
J Surg Res ; 163(2): 317-22, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20599210

RESUMEN

BACKGROUND: Current blood banking practices allow fresh frozen plasma (FFP) to be thawed and then stored for 5 d between 1 and 6 °C. We hypothesized that aged plasma (d 5 FFP) would be pro-inflammatory to the endothelium compared with fresh plasma (d 0 FFP). MATERIALS AND METHODS: Human pulmonary endothelial cells (PECs) were treated with (1) media, (2) media + lipopolysaccharide (LPS), (3) lactated Ringer's (LR), (4) LR + LPS, (5) d 0 FFP, (6) d 0 FFP + LPS, (7) d 5 FFP, and (8) d 5 FFP + LPS. After a 24 h incubation, the PECs were stained with antibodies for I-CAM, V-CAM, P-selectin, and E-selectin. The cells were subsequently analyzed by flow cytometry. RESULTS: In both PEC groups treated with FFP and stimulated with LPS, I-CAM, V-CAM, P-selectin, and E-selectin were significantly up-regulated compared with LR when stimulated by LPS. CONCLUSION: FFP at both ages significantly increased expression of four different adhesion molecules compared with LR in PECs. This may represent a possible mechanism for increased leukocyte binding on the endothelium as a result of FFP transfusion.


Asunto(s)
Moléculas de Adhesión Celular/análisis , Células Endoteliales/química , Pulmón/química , Plasma/fisiología , Células Cultivadas , Selectina E/análisis , Humanos , Molécula 1 de Adhesión Intercelular/análisis , Lipopolisacáridos/farmacología , Selectina-P/análisis , Síndrome de Dificultad Respiratoria/etiología , Molécula 1 de Adhesión Celular Vascular/análisis
6.
Cell Death Dis ; 11(6): 460, 2020 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-32541677

RESUMEN

Disrupting functional protein homeostasis is an established therapeutic strategy for certain tumors. Ongoing studies are evaluating autophagy inhibition for overcoming chemotherapeutic resistance to such therapies by neutralizing lysosomal pH. New and sensitive methods to monitor autophagy in patients are needed to improve trial design and interpretation. We report that mitochondrial-damaged breast cancer cells and rat breast tumors accumulate p53-positive protein aggregates that resist lysosomal degradation. These aggregates were localized to enzymatically-active autolysosomes that were degrading autophagosomes and the autophagic receptor proteins TAX1BP1 and NDP52. NDP52 was identified to associate with aggregated proteins and knocking down NDP52 led to the accumulation of protein aggregates. TAX1BP1 was identified to partly localize with aggregates, and knocking down TAX1BP1 enhanced aggregate formation, suppressed autophagy, impaired NDP52 autophagic degradation and induced cell death. We propose that quantifying aggregates and autophagic receptors are two potential methods to evaluate autophagy and lysosomal degradation, as confirmed using primary human tumor samples. Collectively, this report establishes protein aggregates and autophagy receptors, TAX1BP1 and NDP52, as potential endpoints for monitoring autophagy during drug development and clinical studies.


Asunto(s)
Neoplasias de la Mama/genética , Lisosomas/metabolismo , Mitocondrias/fisiología , Autofagia , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Humanos
7.
J Am Coll Surg ; 230(1): 26-36.e1, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31672677

RESUMEN

BACKGROUND: Endoscopic ultrasound-guided fine-needle aspiration fails to diagnose up to 25% of patients with pancreatic ductal adenocarcinoma (PDAC). Proteomics can help to overcome this clinical dilemma. We hypothesized that soluble protein signatures can differentiate PDAC from benign tissues. STUDY DESIGN: Tissues were obtained from resected surgical specimens, lysed, and homogenates collected for analysis with a 41-protein multiplex assay. Analyte concentrations were normalized to total protein. Statistical analysis was performed to evaluate for differences in PDAC vs benign tissue. RESULTS: Tissues were obtained from 159 patients, 82 patients with PDAC naïve to therapy and 77 with benign pancreatic pathology. Fourteen analytes had a receiver operating characteristic curve area of >0.75 for predicting PDAC vs benign tissue. A recursive partitioning model using only 2 analytes, interleukin 1 receptor antagonist and transforming growth factor-α, provided an accuracy, sensitivity, and specificity of 91.2%, 90.2%, and 92.2%, respectively. A penalized logistic regression model found 12 analytes that provide diagnostic value to a protein signature. The mean area under the receiver operating characteristic after 50 tenfold cross-validations was 0.951. Accuracy, sensitivity, and specificity of this model were 91.2%, 87.8%, and 94.8%, respectively. Applying the scenario of 80% disease prevalence in patients undergoing endoscopic ultrasound with fine-needle aspiration for a pancreatic head mass, positive predictive value is 98.5% (95% CI 93.0% to 99.7%) and negative predictive value is 66.0% (95% CI 54.9% to 75.6%). CONCLUSIONS: Protein signatures from pancreatic specimens can differentiate PDAC from benign tissue. Additional work to validate these findings in a unique sample set is warranted.


Asunto(s)
Carcinoma Ductal Pancreático/química , Carcinoma Ductal Pancreático/diagnóstico , Proteínas de Neoplasias/análisis , Neoplasias Pancreáticas/química , Neoplasias Pancreáticas/diagnóstico , Proteómica , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos
8.
Cancers (Basel) ; 12(2)2020 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-32024069

RESUMEN

Smoking is highly associated with pancreatic cancer. Nicotine, the addictive component of tobacco, is involved in pancreatic cancer tumorigenesis, metastasis, and chemoresistance. This work aimed to describe the role of nicotine within the pancreatic cancer tumor microenvironment. Nicotine treatment was used in vitro to assess its effect on tumor-associated stromal cells and pancreatic cancer cells. Nicotine treatment was then used in a pancreatic cancer patient-derived xenograft model to study the effects in vivo. Nicotine induced secretion of interleukin 8 (IL-8) by tumor-associated stroma cells in an extracellular signal-regulated kinase (ERK)-dependent fashion. The secreted IL-8 and nicotine acted on the pancreatic cancer cell, resulting in upregulation of IL-8 receptor. Nicotine treatment of mice bearing pancreatic cancer patient-derived xenografts had significantly increased tumor mass, increased tumor-free weight loss, and decreased muscle mass. These represent important pathways through which nicotine acts within the tumor microenvironment and worsens pancreatic cancer-induced cachexia, potentially representing future therapeutic targets.

9.
Artículo en Inglés | MEDLINE | ID: mdl-32055795

RESUMEN

Minimally invasive approaches to abdominal surgical procedures have provided superior outcomes when compared to the open approach and thus have become the standard of care. However, minimally invasive pancreatoduodenectomy (MIPD) presents unique difficulties for both laparoscopic and robotic platforms and remains controversial. Ongoing concerns continue about the minimally invasive approach creating meaningful benefit when system-wide data may suggest MIPD results in increased morbidity and mortality during the learning curve. This treatise explores the current state of MIPD, reviewing the volume and quality of data that supports benefit while contrasting the benefits to the unique challenges associated with MIPD that may lead to unacceptable rates of complications and death. We conclude that in a handful of centers, MIPD confers an iterative but not transformative benefit. Significant barriers to the wide-spread acceptance of MIPD are apparent and persist, including: lack of high level data confirming clinical benefit, well defined patient selection criteria, formal education programs that address challenges of the learning curve, and ultimately value.

10.
Oncotarget ; 9(5): 5764-5777, 2018 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-29464032

RESUMEN

The biology of tumor-associated stroma (TAS) in pancreatic ductal adenocarcinoma (PDAC) is not well understood. The paradoxical observation that stroma-depletion strategies lead to progression of PDAC reinforced the need to critically evaluate the functional contribution of TAS in the initiation and progression of PDAC. PDAC and TAS cells are unique in their expression of specific miRNAs, and this specific miRNA expression pattern alters host to tumor microenvironment interactions. Using primary human pancreatic TAS cells and primary xenograft PDAC cells co-culture, we provide evidence of miRNA trafficking and exchanging between TAS and PDAC cells, in a two-way, cell-contact independent fashion, via extracellular vesicles (EVs) transportation. Selective packaging of miRNAs into EVs led to enrichment of stromal specific miR-145 in EVs secreted by TAS cells. Exosomes, but not microvesicles, derived from human TAS cells demonstrated a tumor suppressive role by inducing PDAC cell apoptosis. This effect was mitigated by anti-miR-145 sequences. Our data suggest that TAS-derived miRNAs are delivered to adjacent PDAC cells via exosomes and suppress tumor cell growth. These data highlight that TAS cells secrete exosomes carrying tumor suppressive genetic materials, a possible anti-tumor capacity. Future work of the development of patient-derived exosomes could have therapeutic implications for unresectable PDAC.

11.
JNCI Cancer Spectr ; 2(3): pky043, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30637373

RESUMEN

BACKGROUND: Cancer cachexia is a catabolic condition characterized by skeletal muscle wasting, consequent to tumor burden, which negatively impacts tolerance to cancer therapies and contributes to increased mortality. Partly because of the limited knowledge of the underlying mechanisms of cancer cachexia derived from human studies, however, the ability to therapeutically intervene remains elusive. The purpose of the current study was therefore to better define the phenotype of skeletal muscle obtained from patients with pancreatic ductal adenocarcinoma (PDAC), which has one of the highest rates of cachexia. METHODS: Morphological analyses were performed on rectus abdominis muscle biopsies obtained from resectable PDAC patients undergoing tumor resection surgery (N = 20) and from weight-stable non-cancer control subjects undergoing benign abdominal surgery (N = 16). PDAC patients with a body weight loss of greater than 5% during the previous 6 months were considered cachectic (N = 15). Statistical tests were two sided. RESULTS: Skeletal muscle from cachectic PDAC patients had increased collagen content compared with non-cancer control subjects (1.43% vs 9.66%, P = .0004, Dunn test). Across all PDAC patients, collagen content positively correlated with body weight loss (P = .0016, r = 0.672), was increased in patients with lymph node metastasis (P = .007, Mann-Whitney U test), and was associated with survival on univariate (HR = 1.08, 95% confidence interval [CI] = 1.02 to 1.04, P = .008) and multivariable analyses (HR = 1.08, 95% CI = 1.00 to 1.17, P = .038). Cachectic PDAC patients also displayed increased lipid deposition (2.63% vs 5.72%, P = .042), infiltration of CD68+ macrophages (63.6 cells/mm2 vs 233.8 cells/mm2, P = .0238), calcium deposition (0.21% vs 2.51%, P = .030), and evidence of deficient cellular quality control mechanisms (Mann-Whitney U test). Transcriptional profiling of all patients supported these findings by identifying gene clusters related to wounding, inflammation, and cellular response to TGF-ß upregulated in cachectic PDAC patients compared with non-cancer control subjects. CONCLUSIONS: To our knowledge, this work is the first to demonstrate increased collagen content in cachectic PDAC patients that is associated with poor survival.

12.
J Gastrointest Surg ; 21(9): 1404-1410, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28567575

RESUMEN

OBJECTIVE: We sought to determine if laparoscopic pancreatoduodenectomy (LPD) is a cost-effective alternative to open pancreatoduodenectomy (OPD). METHODS: Hospital cost data, discharge disposition, readmission rates, and readmission costs from periampullary cancer patient cohorts of LPD and OPD were compared. The surgical cohorts over a 40-month period were clinically similar, consisting of 52 and 50 patients in the LPD and OPD groups, respectively. RESULTS: The total operating room costs were higher in the LPD group as compared to the OPD group (median US$12,290 vs US$11,299; P = 0.05) due to increased costs for laparoscopic equipment and regional nerve blocks (P ≤ 0.0001). Although hospital length of stay was shorter in the LPD group (median 7 vs 8 days; P = 0.025), the average hospital cost was not significantly decreased compared to the OPD group (median $28,496 vs $28,623). Surgery-related readmission rates and associated costs did not differ between groups. Compared to OPD patients, significantly more LPD patients were discharged directly home rather than to other healthcare facilities (88% vs 72%; P = 0.047). CONCLUSION: For the index hospitalization, the cost of LPD is equivalent to OPD. Total episode-of-care costs may favor LPD via reduced post-hospital needs for skilled nursing and rehabilitation.


Asunto(s)
Adenocarcinoma/cirugía , Ampolla Hepatopancreática , Neoplasias Duodenales/cirugía , Laparoscopía/economía , Pancreaticoduodenectomía/economía , Readmisión del Paciente/economía , Análisis Costo-Beneficio , Costos de Hospital/estadística & datos numéricos , Humanos , Tiempo de Internación/economía , Tiempo de Internación/estadística & datos numéricos , Pancreaticoduodenectomía/métodos , Alta del Paciente/estadística & datos numéricos , Readmisión del Paciente/estadística & datos numéricos , Estudios Retrospectivos
13.
Pancreas ; 46(6): 813-819, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28609371

RESUMEN

OBJECTIVE: Limitations associated with current animal models serve as a major obstacle to reliable preclinical evaluation of therapies in pancreatic cancer (PC). In an effort to develop more reliable preclinical models, we have recently established a subcutaneous patient-derived xenograft (PDX) model. However, critical aspects of PC responsible for its highly lethal nature, such as the development of distant metastasis and cancer cachexia, remain underrepresented in the flank PDX model. The purpose of this study was to evaluate the degree to which an orthotopic PDX model of PC recapitulates these aspects of the human disease. METHODS: Human PDX-derived PC tumors were implanted directly into the pancreas of NOD.Cg-Prkdc Il2rg/SzJ mice. Tumor growth, metastasis, and muscle wasting were then evaluated. RESULTS: Orthotopically implanted PDX-derived tumors consistently incorporated into the murine pancreatic parenchyma, metastasized to both the liver and lungs and induced muscle wasting directly proportional to the size of the tumor, consistent of the cancer cachexia syndrome. CONCLUSIONS: Through the orthotopic implantation technique described, we demonstrate a highly reproducible model that recapitulates both local and systemic aspects of human PC.


Asunto(s)
Caquexia/etiología , Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/secundario , Músculo Esquelético/patología , Neoplasias Pancreáticas/complicaciones , Neoplasias Pancreáticas/patología , Animales , Caquexia/patología , Modelos Animales de Enfermedad , Xenoinjertos , Humanos , Ratones Endogámicos NOD , Ratones SCID , Invasividad Neoplásica , Trasplante de Neoplasias , Factores de Tiempo , Carga Tumoral
14.
Cancer Res ; 77(3): 672-683, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27864347

RESUMEN

Cancer cells exert mastery over the local tumor-associated stroma (TAS) to configure protective immunity within the tumor microenvironment. The immunomodulatory character of pancreatic lysates of patients with cancer differs from those with pancreatitis. In this study, we evaluated the cross-talk between pancreatic cancer and its TAS in primary human cell culture models. Upon exposure of TAS to pancreatic cancer cell-conditioned media, we documented robust secretion of IL6 and IL8. This TAS response was MyD88-dependent and sufficient to directly suppress both CD4+ and CD8+ T-cell proliferation, inducing Th17 polarization at the expense of Th1. We found that patients possessed a similar shift in circulating effector memory Th17:Th1 ratios compared with healthy controls. The TAS response also directly suppressed CD8+ T-cell-mediated cytotoxicity. Overall, our results demonstrate how TAS contributes to the production of an immunosuppressive tumor microenvironment in pancreatic cancer. Cancer Res; 77(3); 672-83. ©2016 AACR.


Asunto(s)
Factor 88 de Diferenciación Mieloide/metabolismo , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Escape del Tumor/inmunología , Microambiente Tumoral/inmunología , Separación Celular , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Activación de Linfocitos/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Factor 88 de Diferenciación Mieloide/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor Cross-Talk/fisiología , Células Tumorales Cultivadas
15.
Oncotarget ; 8(1): 1177-1189, 2017 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-27901481

RESUMEN

Cancer cachexia represents a debilitating syndrome that diminishes quality of life and augments the toxicities of conventional treatments. Cancer cachexia is particularly debilitating in patients with pancreatic cancer (PC). Mechanisms responsible for cancer cachexia are under investigation and are largely derived from observations in syngeneic murine models of cancer which are limited in PC. We evaluate the effect of human PC cells on both muscle wasting and the systemic inflammatory milieu potentially contributing to PC-associated cachexia. Specifically, human PC xenografts were generated by implantation of pancreatic cancer cells, L3.6pl and PANC-1, either in the flank or orthotopically within the pancreas. Mice bearing orthotopic xenografts demonstrated significant muscle wasting and atrophy-associated gene expression changes compared to controls. Further, despite the absence of adaptive immunity, splenic tissue from orthotopically engrafted mice demonstrated elevations in several pro-inflammatory cytokines associated with cancer cachexia, including TNFα, IL1ß, IL6 and KC (murine IL8 homologue), when compared to controls. Therefore, data presented here support further investigation into the complexity of cancer cachexia in PC to identify potential targets for this debilitating syndrome.


Asunto(s)
Caquexia/etiología , Caquexia/patología , Neoplasias Pancreáticas/complicaciones , Animales , Caquexia/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Xenoinjertos , Humanos , Mediadores de Inflamación/metabolismo , Ratones , Atrofia Muscular/genética , Atrofia Muscular/metabolismo , Atrofia Muscular/patología , Factores de Transcripción , Microambiente Tumoral/genética
16.
J Biomed Mater Res A ; 103(1): 170-84, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24616366

RESUMEN

Given the central role of dendritic cells (DCs) in directing T-cell phenotypes, the ability of biomaterial-treated DCs to dictate autologous T-cell phenotype was investigated. In this study, we demonstrate that differentially biomaterial-treated DCs differentially directed autologous T-cell phenotype and polarization, depending on the biomaterial used to pretreat the DCs. Immature DCs (iDCs) were derived from human peripheral blood monocytes and treated with biomaterial films of alginate, agarose, chitosan, hyaluronic acid, or 75:25 poly(lactic-co-glycolic acid) (PLGA), followed by co-culture of these biomaterial-treated DCs and autologous T cells. When autologous T cells were co-cultured with DCs treated with biomaterial film/antigen (ovalbumin, OVA) combinations, different biomaterial films induced differential levels of T-cell marker (CD4, CD8, CD25, CD69) expression, as well as differential cytokine profiles [interferon (IFN)-γ, interleukin (IL)-12p70, IL-10, IL-4] in the polarization of T helper (Th) types. Dendritic cells treated with agarose films/OVA induced CD4+CD25+FoxP3+ (T regulatory cells) expression, comparable to untreated iDCs, on autologous T cells in the DC-T co-culture system. Furthermore, in this co-culture, agarose treatment induced release of IL-12p70 and IL-10 at higher levels as compared with DC treatment with other biomaterial films/OVA, suggesting Th1 and Th2 polarization, respectively. Dendritic cells treated with PLGA film/OVA treatment induced release of IFN-γ at higher levels compared with that observed for co-cultures with iDCs or DCs treated with all other biomaterial films. These results indicate that DC treatment with different biomaterial films has potential as a tool for immunomodulation by directing autologous T-cell responses.


Asunto(s)
Materiales Biocompatibles , Células Dendríticas/citología , Inmunofenotipificación , Linfocitos T/citología , Biomarcadores/metabolismo , Polaridad Celular , Técnicas de Cocultivo , Células Dendríticas/inmunología , Humanos
17.
Stem Cells Dev ; 20(1): 89-101, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20446815

RESUMEN

The barrier formed by endothelial cells (ECs) plays an important role in tissue homeostasis by restricting passage of circulating molecules and inflammatory cells. Disruption of the endothelial barrier in pathologic conditions often leads to uncontrolled inflammation and tissue damage. An important component of this barrier is adherens junctions, which restrict paracellular permeability. The transmembrane protein vascular endothelial (VE)-cadherin and its cytoplasmic binding partner ß-catenin are major components of functional adherens junctions. We show that mesenchymal stem cells (MSCs) significantly reduce endothelial permeability in cocultured human umbilical vascular endothelial cells (HUVECs) and following exposure to vascular endothelial growth factor, a potent barrier permeability-enhancing agent. When grown in cocultures with HUVECs, MSCs increased VE-cadherin levels and enhanced recruitment of both VE-cadherin and ß-catenin to the plasma membrane. Enhanced membrane localization of ß-catenin was associated with a decrease in ß-catenin-driven gene transcription. Disruption of the VE-cadherin/ß-catenin interaction by overexpressing a truncated VE-cadherin lacking the ß-catenin interacting domain blocked the permeability-stabilizing effect of MSCs. Interestingly, a conditioned medium from HUVEC-MSC cocultures, but not from HUVEC or MSC cells cultured alone, significantly reduced endothelial permeability. In addition, intravenous administration of MSCs to brain-injured rodents reduced injury-induced enhanced blood-brain barrier permeability. Similar to the effect on in vitro cultures, this stabilizing effect on blood-brain barrier function was associated with increased expression of VE-cadherin. Taken together, these results identify a putative mechanism by which MSCs can modulate vascular EC permeability. Further, our results suggest that the mediator(s) of these vascular protective effects is a secreted factor(s) released as a result of direct MSC-EC interaction.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Permeabilidad Capilar , Endotelio Vascular/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Transducción de Señal , beta Catenina/metabolismo , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/patología , Lesiones Encefálicas/patología , Lesiones Encefálicas/terapia , Permeabilidad Capilar/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Técnicas de Cocultivo , Medios de Cultivo Condicionados/farmacología , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Endotelio Vascular/efectos de los fármacos , Humanos , Inyecciones Intravenosas , Masculino , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Unión Proteica/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Venas Umbilicales/citología
18.
PLoS One ; 6(9): e25171, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21980392

RESUMEN

Hemorrhagic shock (HS) and trauma is currently the leading cause of death in young adults worldwide. Morbidity and mortality after HS and trauma is often the result of multi-organ failure such as acute lung injury (ALI) and acute respiratory distress syndrome (ARDS), conditions with few therapeutic options. Bone marrow derived mesenchymal stem cells (MSCs) are a multipotent stem cell population that has shown therapeutic promise in numerous pre-clinical and clinical models of disease. In this paper, in vitro studies with pulmonary endothelial cells (PECs) reveal that conditioned media (CM) from MSCs and MSC-PEC co-cultures inhibits PEC permeability by preserving adherens junctions (VE-cadherin and ß-catenin). Leukocyte adhesion and adhesion molecule expression (VCAM-1 and ICAM-1) are inhibited in PECs treated with CM from MSC-PEC co-cultures. Further support for the modulatory effects of MSCs on pulmonary endothelial function and inflammation is demonstrated in our in vivo studies on HS in the rat. In a rat "fixed volume" model of mild HS, we show that MSCs administered IV potently inhibit systemic levels of inflammatory cytokines and chemokines in the serum of treated animals. In vivo MSCs also inhibit pulmonary endothelial permeability and lung edema with concurrent preservation of the vascular endothelial barrier proteins: VE-cadherin, Claudin-1, and Occludin-1. Leukocyte infiltrates (CD68 and MPO positive cells) are also decreased in lungs with MSC treatment. Taken together, these data suggest that MSCs, acting directly and through soluble factors, are potent stabilizers of the vascular endothelium and inflammation. These data are the first to demonstrate the therapeutic potential of MSCs in HS and have implications for the potential use of MSCs as a cellular therapy in HS-induced lung injury.


Asunto(s)
Células de la Médula Ósea/citología , Células Endoteliales/citología , Pulmón/patología , Células Madre Mesenquimatosas/citología , Choque Hemorrágico/terapia , Animales , Antígenos CD/metabolismo , Células de la Médula Ósea/efectos de los fármacos , Cadherinas/metabolismo , Adhesión Celular/fisiología , Línea Celular , Células Cultivadas , Quimiocina CCL3/metabolismo , Medios de Cultivo Condicionados/farmacología , Citometría de Flujo , Humanos , Inmunohistoquímica , Molécula 1 de Adhesión Intercelular/metabolismo , Interleucina-10/metabolismo , Leucocitos/metabolismo , Pulmón/metabolismo , Masculino , Células Madre Mesenquimatosas/fisiología , Ratas , Ratas Sprague-Dawley , Choque Hemorrágico/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , beta Catenina/metabolismo
19.
Tissue Eng Part C Methods ; 16(5): 989-97, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20001789

RESUMEN

INTRODUCTION: Current mesenchymal stromal cell (MSC) delivery methods require infusion/implantation through needles and/or catheters. Little investigation into the effect of delivery via catheter injection has been completed. We hypothesize that injection of rat and human MSCs through various clinically relevant-sized catheters and flow rates will not affect cell viability, characterization, or function. METHODS: Both rat and human MSCs were injected through 20-, 25-, and 30-gauge needles, as well through an SL-10 microcatheter at rates of 60, 120, 240, and 500 mL/h. MSC viability and apoptotic fraction was measured. MSCs were characterized 24 h after injection with flow cytometric immunophenotyping, and multilineage differentiation was completed. RESULTS: Catheter diameter or flow rate did not affect rat MSC viability. No clinically significant decrease in human MSC viability was observed immediately after injection; however, a delayed decrease in viability was observed at 24 h. No difference in the surface markers CD11b, CD45, CD29, CD49e, CD73, CD90, CD105, and Stro-1 or the capacity for multilineage differentiation (adipogenesis, osteogenesis, and chondrogenesis) was observed for either rat or human MSCs. CONCLUSION: The injection of human and rat MSCs through various clinically relevant catheters and flow rates did not have a clinically significant effect on viability immediately after injection, indicating compliance with recently published Food and Drug Administration guidelines (viability >70%). Further, no changes in cell characterization or function were observed via measurement of cell surface markers and the capacity for multilineage differentiation, respectively. These results ensure the biocompatibility of MSCs with commonly used delivery methods.


Asunto(s)
Supervivencia Celular , Mesodermo/citología , Agujas , Células del Estroma/química , Animales , Apoptosis , Diferenciación Celular , Trasplante de Células , Humanos , Inmunofenotipificación , Mesodermo/inmunología , Ratas , Ratas Sprague-Dawley , Células del Estroma/inmunología
20.
Exp Neurol ; 225(2): 341-52, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20637752

RESUMEN

Recent investigation has shown an interaction between transplanted progenitor cells and resident splenocytes leading to the modulation of the immunologic response in neurological injury. We hypothesize that the intravenous injection of multipotent adult progenitor cells (MAPC) confers neurovascular protection after traumatic brain injury through an interaction with resident splenocytes, subsequently leading to preservation of the blood brain barrier. Four groups of rats underwent controlled cortical impact injury (3 groups) or sham injury (1 group). MAPC were injected via the tail vein at two doses (2*10(6) MAPC/kg or 10*10(6) MAPC/kg) 2 and 24h after injury. Blood brain barrier permeability was assessed by measuring Evans blue dye extravasation (n=6/group). Additionally, splenic mass was measured (n=12/group) followed by splenocyte characterization (n=9/group) including: cell cycle analysis (n=6/group), apoptosis index (n=6/group), cell proliferation (n=6/group), and inflammatory cytokine measurements (n=6/group). Vascular architecture was determined by immunohistochemistry (n=3/group). Traumatic brain injury results in a decrease in splenic mass and increased blood brain barrier permeability. Intravenous infusion of MAPC preserved splenic mass and returned blood brain barrier permeability towards control sham injured levels. Splenocyte characterization indicated an increase in the number and proliferative rate of CD4+ T cells as well as an increase in IL-4 and IL-10 production in stimulated splenocytes isolated from the MAPC treatment groups. Immunohistochemistry demonstrated stabilization of the vascular architecture in the peri-lesion area. Traumatic brain injury causes a reduction in splenic mass that correlates with an increase in circulating immune cells leading to increased blood brain barrier permeability. The intravenous injection of MAPC preserves splenic mass and the integrity of the blood brain barrier. Furthermore, the co-localization of transplanted MAPC and resident CD4+ splenocytes is associated with a global increase in IL-4 and IL-10 production and stabilization of the cerebral microvasculature tight junction proteins.


Asunto(s)
Células Madre Adultas/trasplante , Barrera Hematoencefálica/metabolismo , Lesiones Encefálicas/terapia , Bazo/citología , Trasplante de Células Madre/métodos , Análisis de Varianza , Animales , Barrera Hematoencefálica/patología , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Linfocitos T CD4-Positivos/citología , Recuento de Células , Proliferación Celular , Células Cultivadas , Inmunohistoquímica , Inflamación/metabolismo , Inflamación/patología , Infusiones Intravenosas , Interleucina-10/metabolismo , Interleucina-4/metabolismo , Masculino , Tamaño de los Órganos , Permeabilidad , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Bazo/metabolismo , Bazo/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA