Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 29
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Methods Mol Biol ; 2592: 175-184, 2023.
Article de Anglais | MEDLINE | ID: mdl-36507993

RÉSUMÉ

Innovations in the field of amphiphilic block copolymers have led to the development of a series of attractive polymer-based drug and gene delivery micellar formulations. The amphiphilic block copolymers' low critical micelle concentration (CMC) results in highly stable nanoscale micelles possessing favorable in vivo safety profiles and biocompatibility, making them an excellent carrier choice for the systemic administration of various poorly soluble drugs. These micelles can also be used as an actively targeted drug delivery system. The targeting is achieved by conjugating specific targeting ligand molecules to the micelle surface. The conjugation takes place at the hydrophilic termini of the copolymers, which forms the shell or surface of the nanomicelles. In our lab, we have developed a targeted Pluronic® F127-based nanoformulation to achieve targeting of specific cell types in the pancreas. To achieve active targeting based on the desired end application, we have conjugated several monoclonal antibodies (~150 kDa IgG) reactive to specific cell types in the pancreas by coupling lysine amino groups of the antibody to the p-nitrophenyl carbonate groups generated on the hydrophilic PEO segments of the Pluronic® F127. The resultant targeted nanomicelles demonstrated high binding specificity and targeting efficiency. These nanomicelles can be used to encapsulate and deliver hydrophobic imaging agents and/or water-insoluble therapeutic small molecules to specific pancreatic cell types, enabling the development of diverse tools for use in the diagnosis and/or treatment of pancreas pathologies.


Sujet(s)
Vecteurs de médicaments , Micelles , Vecteurs de médicaments/composition chimique , Systèmes de délivrance de médicaments/méthodes , Polymères/composition chimique , Poloxamère , Pancréas
2.
Sci Rep ; 12(1): 19127, 2022 11 09.
Article de Anglais | MEDLINE | ID: mdl-36351917

RÉSUMÉ

Type 1 diabetes (T1D) occurs as a consequence of the autoimmune destruction of insulin-producing pancreatic beta (ß) cells and commonly presents with insulin deficiency and unregulated glycemic control. Despite improvements in the medical management of T1D, life-threatening complications are still common. Beta-cell replication to replace lost cells may be achieved by using small-molecule mitogenic drugs, like harmine. However, the safe and effective delivery of such drugs to beta cells remains a challenge. This work aims to deploy an antibody conjugated nanocarrier platform to achieve cell-specific delivery of candidate therapeutic and imaging agents to pancreatic endocrine cells. We approached this goal by generating core-shell type micellar nanocarriers composed of the tri-block copolymer, Pluronic®F127 (PEO100-PPO65-PEO100). We decorated these nanocarriers with a pancreatic endocrine cell-selective monoclonal antibody (HPi1), with preference for beta cells, to achieve active targeting. The PPO-based hydrophobic core allows encapsulation of various hydrophobic cargoes, whereas the PEO-based hydrophilic shell curbs the protein adhesion, hence prolonging the nanocarriers' systemic circulation time. The nancarriers were loaded with quantum dots (QDots) that allowed nanocarrier detection both in-vitro and in-vivo. In-vitro studies revealed that HPi1 conjugated nanocarriers could target endocrine cells in dispersed islet cell preparations with a high degree of specificity, with beta cells exhibiting a fluorescent quantum dot signal that was approximately five orders of magnitude greater than the signal associated with alpha cells. In vivo endocrine cell targeting studies demonstrated that the HPi1 conjugated nanocarriers could significantly accumulate at the islet xenograft site. For drug delivery studies, the nanocarriers were loaded with harmine. We demonstrated that HPi1 conjugated nanocarriers successfully targeted and delivered harmine to human endocrine cells in a human islet xenograft model. In this model, targeted harmine delivery yielded an ~ 41-fold increase in the number of BrdU positive cells in the human islet xenograft than that observed in untreated control mice. By contrast, non-targeted harmine yielded an ~ 9-fold increase in BrdU positive cells. We conclude that the nanocarrier platform enabled cell-selective targeting of xenografted human pancreatic endocrine cells and the selective delivery of the hydrophobic drug harmine to those cells. Further, the dramatic increase in proliferation with targeted harmine, a likely consequence of achieving higher local drug concentrations, supports the concept that targeted drug delivery may promote more potent biological responses when using harmine and/or other drugs than non-targeting approaches. These results suggest that this targeted drug delivery platform may apply in drug screening, beta cell regenerative therapies, and/or diagnostic imaging in patients with type 1 diabetes.


Sujet(s)
Diabète de type 1 , Ilots pancréatiques , Humains , Souris , Animaux , Harmine/pharmacologie , Hétérogreffes , Broxuridine , Ilots pancréatiques/métabolisme , Prolifération cellulaire , Insuline/métabolisme
3.
Stem Cell Reports ; 17(4): 979-992, 2022 04 12.
Article de Anglais | MEDLINE | ID: mdl-35245441

RÉSUMÉ

Cell replacement therapy using ß cells derived from stem cells is a promising alternative to conventional diabetes treatment options. Although current differentiation methods produce glucose-responsive ß cells, they can also yield populations of undesired endocrine progenitors and other proliferating cell types that might interfere with long-term islet function and safety of transplanted cells. Here, we describe the generation of an array of monoclonal antibodies against cell surface markers that selectively label stem cell-derived islet cells. A high-throughput screen identified promising candidates, including three clones that mark a high proportion of endocrine cells in differentiated cultures. A scalable magnetic sorting method was developed to enrich for human pluripotent stem cell (hPSC)-derived islet cells using these three antibodies, leading to the formation of islet-like clusters with improved glucose-stimulated insulin secretion and reduced growth upon transplantation. This strategy should facilitate large-scale production of functional islet clusters from stem cells for disease modeling and cell replacement therapy.


Sujet(s)
Cellules à insuline , Ilots pancréatiques , Cellules souches pluripotentes , Différenciation cellulaire , Glucose/métabolisme , Humains , Insuline/métabolisme , Sécrétion d'insuline , Cellules à insuline/métabolisme , Cellules souches pluripotentes/métabolisme
5.
Article de Anglais | MEDLINE | ID: mdl-29928673

RÉSUMÉ

BACKGROUND & AIMS: Continual renewal of the intestinal epithelium is dependent on active- and slow-cycling stem cells that are confined to the crypt base. Tight regulation of these stem cell populations maintains homeostasis by balancing proliferation and differentiation to support critical intestinal functions. The hierarchical relation of discrete stem cell populations in homeostasis or during regenerative epithelial repair remains controversial. Although recent studies have supported a model for the active-cycling leucine-rich repeat-containing G-protein-coupled receptor 5 (Lgr5)+ intestinal stem cell (ISC) functioning upstream of the slow-cycling B lymphoma Mo-MLV insertion region 1 homolog (Bmi1)-expressing cell, other studies have reported the opposite relation. Tools that facilitate simultaneous analyses of these populations are required to evaluate their coordinated function. METHODS: We used novel monoclonal antibodies (mAbs) raised against murine intestinal epithelial cells in conjunction with ISC-green fluorescent protein (GFP) reporter mice to analyze relations between ISC populations by microscopy. Ex vivo 3-dimensional cultures, flow cytometry, and quantitative reverse-transcription polymerase chain reaction analyses were performed. RESULTS: Two novel mAbs recognized distinct subpopulations of the intestinal epithelium and when used in combination permitted isolation of discrete Lgr5GFP and Bmi1GFP-enriched populations with stem activity. Growth from singly isolated Lgr5GFP ISCs gave rise to small spheroids. Spheroids did not express Lgr5GFP and instead up-regulated Bmi1GFP expression. Conversely, Bmi1-derived spheroids initiated Lgr5GFP expression as crypt domains were established. CONCLUSIONS: These data showed the functional utility of murine mAbs in the isolation and investigation of Lgr5GFP and Bmi1GFP ISC-enriched populations. Ex vivo analyses showed hierarchical plasticity between different ISC-expressing states; specifically Lgr5GFP ISCs gave rise to Bmi1GFP cells, and vice versa. These data highlight the impact of temporal and physiological context on unappreciated interactions between Lgr5GFP and Bmi1GFP cells during crypt formation.

6.
J Immunol Methods ; 459: 20-28, 2018 08.
Article de Anglais | MEDLINE | ID: mdl-29758224

RÉSUMÉ

We describe here the humanisation of two mouse monoclonal antibodies that bind to surface markers on human pancreatic islet endocrine cells. Monoclonal antibodies produced by the HIC1-2B4 and HIC0-4F9 mouse hybridomas bind distinct surface molecules expressed on endocrine cells and have been validated for a number of experimental methods including immunohistochemistry and live cell sorting by flow cytometry. Variable region framework and first constant region domain sequences were replaced with that from compatible human antibody sequences, and the resulting recombinant antigen-binding fragments were cloned and expressed in mouse myeloma cells. ELISA, fluorescent immunohistochemistry, and flow cytometry were used to assess the specificity of the humanised antibody fragments. Purification and binding analyses indicated that human islet endocrine cell binding was retained in the humanised antibody fragments. These humanised, recombinant antibody fragments have a lower risk of eliciting adverse responses from a patient's immune system and, therefore, have highly improved clinical potential. Thus, they may be used to image, target or carry cargo specifically to islet cells in human patients.


Sujet(s)
Anticorps monoclonaux humanisés/métabolisme , Fragments Fab d'immunoglobuline/métabolisme , Ilots pancréatiques/immunologie , Ilots pancréatiques/métabolisme , Animaux , Anticorps monoclonaux humanisés/isolement et purification , Spécificité des anticorps , Sites de fixation des anticorps , Test ELISA , Cytométrie en flux , Humains , Hybridomes , Fragments Fab d'immunoglobuline/isolement et purification , Immunohistochimie , Souris , Modèles moléculaires , Protéines recombinantes/immunologie , Protéines recombinantes/métabolisme
7.
Am J Physiol Renal Physiol ; 314(4): F501-F516, 2018 04 01.
Article de Anglais | MEDLINE | ID: mdl-29187371

RÉSUMÉ

The erythropoietin receptor (EpoR) is widely expressed but its renoprotective action is unexplored. To examine the role of EpoR in vivo in the kidney, we induced acute kidney injury (AKI) by ischemia-reperfusion in mice with different EpoR bioactivities in the kidney. EpoR bioactivity was reduced by knockin of wild-type human EpoR, which is hypofunctional relative to murine EpoR, and a renal tubule-specific EpoR knockout. These mice had lower EPO/EpoR activity and lower autophagy flux in renal tubules. Upon AKI induction, they exhibited worse renal function and structural damage, more apoptosis at the acute stage (<7 days), and slower recovery with more tubulointerstitial fibrosis at the subacute stage (14 days). In contrast, mice with hyperactive EpoR signaling from knockin of a constitutively active human EpoR had higher autophagic flux, milder kidney damage, and better renal function at the acute stage but, surprisingly, worse tubulointerstitial fibrosis and renal function at the subacute stage. Either excess or deficient EpoR activity in the kidney was associated with abnormal peritubular capillaries and tubular hypoxia, creating a "U-shaped" relationship. The direct effects of EpoR on tubular cells were confirmed in vitro by a hydrogen peroxide model using primary cultured proximal tubule cells with different EpoR activities. In summary, normal erythropoietin (EPO)/EpoR signaling in renal tubules provides defense against renal tubular injury maintains the autophagy-apoptosis balance and peritubular capillary integrity. High and low EPO/EpoR bioactivities both lead to vascular defect, and high EpoR activity overides the tubular protective effects in AKI recovery.


Sujet(s)
Atteinte rénale aigüe/métabolisme , Vaisseaux capillaires/métabolisme , Érythropoïétine/métabolisme , Tubules contournés proximaux/vascularisation , Tubules contournés proximaux/métabolisme , Néovascularisation physiologique , Récepteur érythropoïétine/métabolisme , Atteinte rénale aigüe/génétique , Atteinte rénale aigüe/anatomopathologie , Atteinte rénale aigüe/physiopathologie , Animaux , Apoptose , Autophagie , Vaisseaux capillaires/anatomopathologie , Vaisseaux capillaires/physiopathologie , Hypoxie cellulaire , Cellules cultivées , Modèles animaux de maladie humaine , Fibrose , Humains , Tubules contournés proximaux/anatomopathologie , Tubules contournés proximaux/physiopathologie , Souris de souche-129 , Souris transgéniques , Récepteur érythropoïétine/déficit , Récepteur érythropoïétine/génétique , Transduction du signal
8.
Nat Commun ; 8(1): 331, 2017 08 24.
Article de Anglais | MEDLINE | ID: mdl-28835709

RÉSUMÉ

PDX1+/NKX6-1+ pancreatic progenitors (PPs) give rise to endocrine cells both in vitro and in vivo. This cell population can be successfully differentiated from human pluripotent stem cells (hPSCs) and hold the potential to generate an unlimited supply of ß cells for diabetes treatment. However, the efficiency of PP generation in vitro is highly variable, negatively impacting reproducibility and validation of in vitro and in vivo studies, and consequently, translation to the clinic. Here, we report the use of a proteomics approach to phenotypically characterize hPSC-derived PPs and distinguish these cells from non-PP populations during differentiation. Our analysis identifies the pancreatic secretory granule membrane major glycoprotein 2 (GP2) as a PP-specific cell surface marker. Remarkably, GP2 is co-expressed with NKX6-1 and PTF1A in human developing pancreata, indicating that it marks the multipotent pancreatic progenitors in vivo. Finally, we show that isolated hPSC-derived GP2+ cells generate ß-like cells (C-PEPTIDE+/NKX6-1+) more efficiently compared to GP2- and unsorted populations, underlining the potential therapeutic applications of GP2.Pancreatic progenitors (PPs) can be derived from human pluripotent stem cells in vitro but efficiency of differentiation varies, making it hard to sort for insulin-producing cells. Here, the authors use a proteomic approach to identify the secretory granule membrane glycoprotein 2 as a marker for PDX1+/NKX6-1+ PPs.


Sujet(s)
Marqueurs biologiques tumoraux/métabolisme , Membrane cellulaire/métabolisme , Pancréas/métabolisme , Cellules souches/métabolisme , Différenciation cellulaire , Cellules cultivées , Protéines liées au GPI , Protéines à homéodomaine/métabolisme , Humains , Cellules à insuline/métabolisme , Spectrométrie de masse , Pancréas/cytologie , Cellules souches pluripotentes/cytologie , Cellules souches pluripotentes/métabolisme , Protéomique/méthodes , Transactivateurs/métabolisme , Facteurs de transcription/métabolisme
9.
Nat Commun ; 7: 11756, 2016 07 11.
Article de Anglais | MEDLINE | ID: mdl-27399229

RÉSUMÉ

Human pancreatic islets of Langerhans contain five distinct endocrine cell types, each producing a characteristic hormone. The dysfunction or loss of the insulin-producing ß cells causes diabetes mellitus, a disease that harms millions. Until now, ß cells were generally regarded as a single, homogenous cell population. Here we identify four antigenically distinct subtypes of human ß cells, which we refer to as ß1-4, and which are distinguished by differential expression of ST8SIA1 and CD9. These subpopulations are always present in normal adult islets and have diverse gene expression profiles and distinct basal and glucose-stimulated insulin secretion. Importantly, the ß cell subtype distribution is profoundly altered in type 2 diabetes. These data suggest that this antigenically defined ß cell heterogeneity is functionally and likely medically relevant.


Sujet(s)
Diabète de type 2/métabolisme , Cellules à insuline/cytologie , Sialyltransferases/métabolisme , Antigène CD9/métabolisme , Adulte , Sujet âgé , Femelle , Cytométrie en flux , Hémoglobine glyquée/métabolisme , Humains , Cellules à insuline/métabolisme , Ilots pancréatiques , Mâle , Adulte d'âge moyen , Jeune adulte
10.
Development ; 142(24): 4253-65, 2015 Dec 15.
Article de Anglais | MEDLINE | ID: mdl-26493401

RÉSUMÉ

The efficient generation of hepatocytes from human pluripotent stem cells (hPSCs) requires the induction of a proper endoderm population, broadly characterized by the expression of the cell surface marker CXCR4. Strategies to identify and isolate endoderm subpopulations predisposed to the liver fate do not exist. In this study, we generated mouse monoclonal antibodies against human embryonic stem cell-derived definitive endoderm with the goal of identifying cell surface markers that can be used to track the development of this germ layer and its specification to a hepatic fate. Through this approach, we identified two endoderm-specific antibodies, HDE1 and HDE2, which stain different stages of endoderm development and distinct derivative cell types. HDE1 marks a definitive endoderm population with high hepatic potential, whereas staining of HDE2 tracks with developing hepatocyte progenitors and hepatocytes. When used in combination, the staining patterns of these antibodies enable one to optimize endoderm induction and hepatic specification from any hPSC line.


Sujet(s)
Marqueurs biologiques/métabolisme , Différenciation cellulaire , Endoderme/cytologie , Hépatocytes/cytologie , Cellules souches pluripotentes/cytologie , Animaux , Anticorps/métabolisme , Lignée cellulaire , Séparation cellulaire , Ectoderme/cytologie , Hépatocytes/métabolisme , Cellules souches embryonnaires humaines/cytologie , Cellules souches embryonnaires humaines/métabolisme , Humains , Cinétique , Mésoderme/cytologie , Souris de lignée BALB C , Pancréas/embryologie , Cellules souches pluripotentes/métabolisme , Coloration et marquage
11.
Stem Cell Res ; 15(1): 172-81, 2015 Jul.
Article de Anglais | MEDLINE | ID: mdl-26079872

RÉSUMÉ

Novel cell surface-reactive monoclonal antibodies generated against extrahepatic biliary cells were developed for the isolation and characterization of different cell subsets from normal adult human gallbladder. Eleven antigenically distinct gallbladder subpopulations were isolated by fluorescence-activated cell sorting. They were classified into epithelial, mesenchymal, and pancreatobiliary (PDX1(+)SOX9(+)) subsets based on gene expression profiling. These antigenically distinct human gallbladder cell subsets could potentially also reflect different functional properties in regards to bile physiology, cell renewal and plasticity. Three of the novel monoclonal antibodies differentially labeled archival sections of primary carcinoma of human gallbladder relative to normal tissue. The novel monoclonal antibodies described herein enable the identification and characterization of antigenically diverse cell subsets within adult human gallbladder and are putative tumor biomarkers.


Sujet(s)
Marqueurs biologiques/métabolisme , Vésicule biliaire/métabolisme , Adénocarcinome/anatomopathologie , Adulte , Animaux , Anticorps/immunologie , Conduits biliaires extrahépatiques/métabolisme , Lignée cellulaire tumorale , Séparation cellulaire , Conduit cystique/métabolisme , Cellules épithéliales/cytologie , Femelle , Cytométrie en flux , Technique d'immunofluorescence , Vésicule biliaire/anatomopathologie , Régulation de l'expression des gènes , Humains , Mésoderme/cytologie , Souris de lignée BALB C , Pancréas/métabolisme , Coloration et marquage
12.
Stem Cell Res ; 13(2): 275-83, 2014 Sep.
Article de Anglais | MEDLINE | ID: mdl-25151611

RÉSUMÉ

Pancreatic Lgr5 expression has been associated with organoid-forming epithelial progenitor populations but the identity of the organoid-initiating epithelial cell subpopulation has remained elusive. Injury causes the emergence of an Lgr5(+) organoid-forming epithelial progenitor population in the adult mouse liver and pancreas. Here, we define the origin of organoid-initiating cells from mouse pancreas and liver prior to Lgr5 activation. This clonogenic population was defined as MIC1-1C3(+)/CD133(+)/CD26(-) in both tissues and the frequency of organoid initiation within this population was approximately 5% in each case. The transcriptomes of these populations overlapped extensively and showed enrichment of epithelial progenitor-associated regulatory genes such as Sox9 and FoxJ1. Surprisingly, pancreatic organoid cells also had the capacity to generate hepatocyte-like cells upon transplantation to Fah(-/-) mice, indicating a differentiation capacity similar to hepatic organoids. Although spontaneous endocrine differentiation of pancreatic progenitors was not observed in culture, adenoviral delivery of fate-specifying factors Pdx1, Neurog3 and MafA induced insulin expression without glucagon or somatostatin. Pancreatic organoid cultures therefore preserve many key attributes of progenitor cells while allowing unlimited expansion, facilitating the study of fate determination.


Sujet(s)
Différenciation cellulaire , Lignage cellulaire , Hépatocytes/métabolisme , Foie/métabolisme , Organoïdes/métabolisme , Pancréas/métabolisme , Cellules souches/métabolisme , Adenoviridae/génétique , Animaux , Marqueurs biologiques/métabolisme , Prolifération cellulaire , Séparation cellulaire/méthodes , Cellules cultivées , Régulation de l'expression des gènes au cours du développement , Vecteurs génétiques , Hépatocytes/transplantation , Hydrolases/déficit , Hydrolases/génétique , Insuline/métabolisme , Foie/cytologie , Souris , Souris de lignée C57BL , Souris knockout , Organoïdes/cytologie , Pancréas/cytologie , Phénotype , Transduction du signal , Transplantation de cellules souches , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Transduction génétique
14.
J Clin Invest ; 123(3): 1275-84, 2013 Mar.
Article de Anglais | MEDLINE | ID: mdl-23434589

RÉSUMÉ

Insulin-secreting ß cells and glucagon-secreting α cells maintain physiological blood glucose levels, and their malfunction drives diabetes development. Using ChIP sequencing and RNA sequencing analysis, we determined the epigenetic and transcriptional landscape of human pancreatic α, ß, and exocrine cells. We found that, compared with exocrine and ß cells, differentiated α cells exhibited many more genes bivalently marked by the activating H3K4me3 and repressing H3K27me3 histone modifications. This was particularly true for ß cell signature genes involved in transcriptional regulation. Remarkably, thousands of these genes were in a monovalent state in ß cells, carrying only the activating or repressing mark. Our epigenomic findings suggested that α to ß cell reprogramming could be promoted by manipulating the histone methylation signature of human pancreatic islets. Indeed, we show that treatment of cultured pancreatic islets with a histone methyltransferase inhibitor leads to colocalization of both glucagon and insulin and glucagon and insulin promoter factor 1 (PDX1) in human islets and colocalization of both glucagon and insulin in mouse islets. Thus, mammalian pancreatic islet cells display cell-type-specific epigenomic plasticity, suggesting that epigenomic manipulation could provide a path to cell reprogramming and novel cell replacement-based therapies for diabetes.


Sujet(s)
Épigenèse génétique , Cellules à glucagon/physiologie , Cellules à insuline/physiologie , Maturation post-traductionnelle des protéines , Animaux , Différenciation cellulaire , Séparation cellulaire , Cellules cultivées , Antienzymes/pharmacologie , Cytométrie en flux , Analyse de profil d'expression de gènes , Cellules à glucagon/métabolisme , Histone méthyltransférases , Histone-lysine N-methyltransferase/antagonistes et inhibiteurs , Histone-lysine N-methyltransferase/métabolisme , Histone/métabolisme , Humains , Cellules à insuline/métabolisme , Méthylation , Souris , Annotation de séquence moléculaire , Analyse de séquence d'ARN , Transcriptome
15.
Cancer Cytopathol ; 121(1): 37-46, 2013 Jan.
Article de Anglais | MEDLINE | ID: mdl-22811080

RÉSUMÉ

BACKGROUND.: Pancreatic ductal adenocarcinoma is rarely detected early enough for patients to be cured. The objective of the authors was to develop a monoclonal antibody to distinguish adenocarcinoma and precancerous intraductal papillary mucinous neoplasia (IPMN) from benign epithelium. METHODS.: Mice were immunized with human pancreatic adenocarcinoma cells and monoclonal antibodies were screened against a panel of archived pancreatic tissue sections, including pancreatitis (23 cases), grade 1 IPMN (16 cases), grade 2 IPMN (9 cases), grade 3 IPMN (13 cases), and various grades of adenocarcinoma (17 cases). One monoclonal antibody, human pancreatic cancer fusion 2 (HPC2) 1-B3, which specifically immunostained adenocarcinoma and all grades of IPMN, was isolated. Subsequently, HPC2 1-B3 was evaluated in a retrospective series of 31 fine-needle aspiration (FNA) biopsies from clinically suspicious pancreatic lesions that had long-term clinical follow-up. RESULTS.: HPC2 1-B3 was negative in all 31 cases of chronic pancreatitis that were tested. In contrast, HPC2 1-B3 immunostained the cytoplasm and luminal surface of all 16 well- to moderately differentiated pancreatic ductal adenocarcinomas. It demonstrated only weak focal staining of poorly differentiated carcinomas. All high-grade IPMNs were found to be positive for HPC2 1-B3. The majority of low-grade to intermediate-grade IPMNs were positive (66% of cases). Immunostaining a separate series of pancreatic FNA cell blocks for HPC2 1-B3 demonstrated that the relative risk for detecting at least low-grade dysplasia (2.0 [95% confidence interval, 1.23-3.26]) was statistically significant (P = .002 by the Fisher exact test). CONCLUSIONS.: To reduce the mortality of pancreatic cancer, more effective early screening methods are necessary. The data from the current study indicate that a novel monoclonal antibody, HPC2 1-B3, may facilitate the diagnosis of early pancreatic dysplasia.


Sujet(s)
Marqueurs biologiques tumoraux/analyse , Carcinome du canal pancréatique/diagnostic , Protéines tumorales/immunologie , Pancréas/anatomopathologie , Tumeurs du pancréas/diagnostic , Adénocarcinome mucineux/anatomopathologie , Animaux , Anticorps monoclonaux , Transformation cellulaire néoplasique , Humains , Hybridomes , Souris , Souris de lignée BALB C , États précancéreux/diagnostic
16.
Hum Pathol ; 43(10): 1583-9, 2012 Oct.
Article de Anglais | MEDLINE | ID: mdl-22406361

RÉSUMÉ

Metastatic pancreatic ductal adenocarcinoma and primary cholangiocarcinoma are morphologically very similar and, therefore, challenging to distinguish in liver biopsies. The distinction is important because surgical management and prognosis differ significantly. Several immunohistochemical markers have been evaluated to aid this diagnosis, but aside from N-cadherin, which labels cholangiocarcinoma, few provide the combination of good sensitivity and specificity. Our laboratory recently developed the novel monoclonal antibody human pancreatic cancer fusion #2 (HPC2) that recognizes pancreatic cancer. We hypothesized that the combination of our new marker and N-cadherin can assist in distinguishing metastatic pancreatic cancer from cholangiocarcinoma. We immunostained resections of 60 pancreatic ductal adenocarcinomas and 31 cholangiocarcinomas for the HPC2 and N-cadherin antigens. We also stained 24 gallbladder adenocarcinomas, 11 ampullary adenocarcinomas, and 10 metastatic colonic adenocarcinomas to the liver. Sections were independently scored by 2 pathologists with good agreement using both markers (κ statistics, 0.62-0.64; P < .0001). HPC2 was observed in 80% of pancreatic cancers (48/60), 82% of ampullary (9/11), and 32% (10/31) of cholangiocarcinomas. N-cadherin stained 27% (16/60) of the pancreas cases and 58% (18/31) of the cholangiocarcinomas. Gallbladder and colon cancers were usually double negative (18/24 and 8/10, respectively). Each marker provided significant likelihood ratios to separate pancreatic cancer (HPC2, 2.48 [1.46-4.19]; P < .0001) from cholangiocarcinoma (N-cadherin, 2.17 [1.3-3.64]; P < .01). The combination of both markers provided even better specificity and positive likelihood ratios. We conclude that HPC2 and N-cadherin significantly improve accurate classification of pancreatic cancer and cholangiocarcinoma.


Sujet(s)
Anticorps monoclonaux , Tumeurs des canaux biliaires/diagnostic , Carcinome du canal pancréatique/diagnostic , Cholangiocarcinome/diagnostic , Tumeurs du pancréas/diagnostic , Tumeurs des canaux biliaires/métabolisme , Conduits biliaires intrahépatiques/anatomopathologie , Marqueurs biologiques tumoraux/analyse , Cadhérines/analyse , Cadhérines/biosynthèse , Carcinome du canal pancréatique/métabolisme , Cholangiocarcinome/métabolisme , Diagnostic différentiel , Humains , Immunohistochimie , Protéines tumorales/analyse , Protéines tumorales/biosynthèse , Tumeurs du pancréas/métabolisme
17.
Nucl Med Biol ; 39(5): 645-51, 2012 Jul.
Article de Anglais | MEDLINE | ID: mdl-22316614

RÉSUMÉ

INTRODUCTION: We previously demonstrated MORF/cMORF pretargeting of human islets and betalox 5 cells (a human beta cell line) transplanted subcutaneously in mice with the anti-human islet antibody, HPi1. We now compare pretargeting with direct targeting in the beta cell transplant model to evaluate the degree to which target/non-target (T/NT) ratios may be improved by pretargeting. METHODS: Specific binding of an anti-human islet antibody HPi1 to the beta cells transplanted subcutaneously in mice was examined against a negative control antibody. We then compared pretargeting by MORF-HPi1 plus 111In-labeled cMORF to direct targeting by 111In-labeled HPi1. RESULTS: HPi1 binding to betalox5 human cells in the transplant was shown by immunofluorescence. Normal organ 111In backgrounds by pretargeting were always lower, although target accumulations were similar. More importantly, the transplant to pancreas and liver ratios was, respectively, 26 and 10 by pretargeting as compared to 9 and 0.6 by direct targeting. CONCLUSIONS: Pretargeting greatly improves the T/NT ratios, and based on the estimated endocrine to exocrine ratio within a pancreas, pretargeting may be approaching the sensitivity required for successful imaging of human islets within this organ.


Sujet(s)
Anticorps/immunologie , Cellules à insuline/immunologie , Cellules à insuline/métabolisme , Morpholinos/métabolisme , Peau , Animaux , Lignée cellulaire , Humains , Radio-isotopes de l'indium , Cellules à insuline/transplantation , Marquage isotopique , Souris , Morpholinos/pharmacocinétique
18.
Mol Cell Endocrinol ; 339(1-2): 144-50, 2011 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-21539888

RÉSUMÉ

Tools permitting the isolation of live pancreatic cell subsets for culture and/or molecular analysis are limited. To address this, we developed a collection of monoclonal antibodies with selective surface labeling of endocrine and exocrine pancreatic cell types. Cell type labeling specificity and cell surface reactivity were validated on mouse pancreatic sections and by gene expression analysis of cells isolated using FACS. Five antibodies which marked populations of particular interest were used to isolate and study viable populations of purified pancreatic ducts, acinar cells, and subsets of acinar cells from whole pancreatic tissue or of alpha or beta cells from isolated mouse islets. Gene expression analysis showed the presence of known endocrine markers in alpha and beta cell populations and revealed that TTR and DPPIV are primarily expressed in alpha cells whereas DGKB and GPM6A have a beta cell specific expression profile.


Sujet(s)
Antigènes de surface/métabolisme , Séparation cellulaire/méthodes , Cellules à glucagon/métabolisme , Cellules à insuline/métabolisme , Conduits pancréatiques/métabolisme , Animaux , Anticorps monoclonaux d'origine murine , Antigènes de surface/immunologie , Diacylglycérol kinase/métabolisme , Dipeptidyl peptidase 4/métabolisme , Cytométrie en flux/méthodes , Glycoprotéines membranaires/métabolisme , Souris , Souris de lignée BALB C , Protéines de tissu nerveux/métabolisme , Pancréas/cytologie , Pancréas/embryologie , Conduits pancréatiques/cytologie , Conduits pancréatiques/embryologie , Préalbumine/métabolisme , Rats , Rats de lignée F344 , Coloration et marquage
19.
Mol Pharm ; 8(3): 767-73, 2011 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-21361360

RÉSUMÉ

Noninvasive measurement of human islet cell mass in pancreas or following islet transplantation by nuclear imaging has yet to be achieved. It has been shown using mouse tumor models that pretargeting imaging strategies are sensitive and can greatly increase target to nontarget signal ratios. The objective now is to demonstrate the specific pretargeting of human islet cells in mice. Our pretargeting strategy uses an anti-human islet cell antibody HPi1, conjugated to a phosphorodiamidate morpholino oligomer (MORF) that binds specifically to a (99m)Tc labeled complementary MORF (cMORF). Sensitivity and specificity of the pretargeting were first validated in culture using a human beta cell line (betalox5) and a negative control human cell line (HEK293). Pretargeting was then used to target and visualize these two cell lines and human islets transplanted subcutaneously in NOD-scid IL2rγ(null) mice. In culture, (99m)Tc accumulation on the betalox5 cells pretargeted by MORF-HPi1 was 100-fold higher than on untreated betalox5 cells or following treatment with native HPi1 and much higher than on the MORF-HPi1 pretargeted control HEK293 cells. Small animal imaging readily localized the transplanted betalox5 cells and human islets, but not the HEK293 cells. Ex vivo counting demonstrated 3-fold higher (99m)Tc accumulation in the transplanted betalox5 cells and human islets than in the control HEK293 cells. The target accumulation was also shown to increase linearly with increased numbers of the implanted betalox5 cells. These results demonstrate specific binding of radioactivity and successful imaging of human betalox5 cells and human islets transplanted in mice. Thus MORF/cMORF pretargeting may be useful to measure noninvasively human islet cell mass within the pancreas or following islet transplantation.


Sujet(s)
Anticorps/métabolisme , Cellules à insuline/cytologie , Cellules à insuline/métabolisme , Cellules à insuline/transplantation , Morpholines/métabolisme , Animaux , Chromatographie en phase liquide à haute performance , Cellules HEK293 , Humains , Souris , Souris de lignée NOD , Souris SCID , Morpholinos
20.
Am J Sports Med ; 38(7): 1324-33, 2010 Jul.
Article de Anglais | MEDLINE | ID: mdl-20423988

RÉSUMÉ

BACKGROUND: Donor-site morbidity, limited numbers of cells, loss of phenotype during ex vivo expansion, and age-related decline in chondrogenic activity present critical obstacles to the use of autologous chondrocyte implantation for cartilage repair. Chondrocytes from juvenile cadaveric donors may represent an alternative to autologous cells. Hypothesis/ PURPOSE: The authors hypothesized that juvenile chondrocyte would show stronger and more stable chondrogenic activity than adult cells in vitro and that juvenile cells pose little risk of immunologic incompatibility in adult hosts. STUDY DESIGN: Controlled laboratory study. METHODS: Cartilage samples were from juvenile (<13 years old) and adult (>13 years old) donors. The chondrogenic activity of freshly isolated human articular chondrocytes and of expanded cells after monolayer culture was measured by proteoglycan assay, gene expression analysis, and histology. Lymphocyte proliferation assays were used to assess immunogenic activity. RESULTS: Proteoglycan content in neocartilage produced by juvenile chondrocytes was 100-fold higher than in neocartilage produced by adult cells. Collagen type II and type IX mRNA in fresh juvenile chondrocytes were 100- and 700-fold higher, respectively, than in adult chondrocytes. The distributions of collagens II and IX were similar in native juvenile cartilage and in neocartilage made by juvenile cells. Juvenile cells grew significantly faster in monolayer cultures than adult cells (P = .002) and proteoglycan levels produced in agarose culture was significantly higher in juvenile cells than in adult cells after multiple passages (P < .001). Juvenile chondrocytes did not stimulate lymphocyte proliferation. CONCLUSION: These results document a dramatic age-related decline in human chondrocyte chondrogenic potential and show that allogeneic juvenile chondrocytes do not stimulate an immunologic response in vivo. CLINICAL RELEVANCE: Juvenile human chondrocytes have greater potential to restore articular cartilage than adult cells, and may be transplanted without the fear of rejection, suggesting a new allogeneic approach to restoring articular cartilage in older individuals.


Sujet(s)
Cartilage articulaire/physiologie , Chondrocytes/physiologie , Chondrocytes/transplantation , Régénération , Adolescent , Adulte , Facteurs âges , Sujet âgé , Maladies du cartilage/chirurgie , Cartilage articulaire/cytologie , Cartilage articulaire/chirurgie , Différenciation cellulaire , Cellules cultivées , Chondrocytes/composition chimique , Collagène de type II/analyse , Collagène de type II/métabolisme , Collagène de type IX/analyse , Collagène de type IX/métabolisme , Femelle , Humains , Nourrisson , Activation des lymphocytes , Mâle , Adulte d'âge moyen , Protéoglycanes/analyse , Protéoglycanes/métabolisme , Jeune adulte
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...