Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
FEBS Open Bio ; 12(10): 1717-1728, 2022 10.
Article in English | MEDLINE | ID: mdl-36062328

ABSTRACT

We previously observed that genomic loss of galectin-3 (Gal-3; encoded by Lgals3) in mice has a significant protective effect on age-related bone loss. Gal-3 has both intracellular and extracellular functionality, and we wanted to assess whether the affect we observed in the Lgals3 knockout (KO) mice could be attributed to the ability of Gal-3 to bind glycoproteins. Mutation of a highly conserved arginine to a serine in human Gal-3 (LGALS3-R186S) blocks glycan binding and secretion. We generated mice with the equivalent mutation (Lgals3-R200S) and observed a subsequent reduction in Gal-3 secretion from mouse embryonic fibroblasts and in circulating blood. When examining bone structure in aged mice, we noticed some similarities to the Lgals3-KO mice and some differences. First, we observed greater bone mass in Lgals3-R200S mutant mice, as was previously observed in Lgals3-KO mice. Like Lgals3-KO mice, significantly increased trabecular bone mass was only observed in female Lgals3-R200S mice. These results suggest that the greater bone mass observed is driven by the loss of extracellular Gal-3 functionality. However, the results from our cortical bone expansion data showed a sex-dependent difference, with only male Lgals3-KO mice having an increased response, contrasting with our earlier study. These notable sex differences suggest a potential role for sex hormones, most likely androgen signaling, being involved. In summary, our results suggest that targeting extracellular Gal-3 function may be a suitable treatment for age-related loss of bone mass.


Subject(s)
Cancellous Bone , Cortical Bone , Galectin 3 , Androgens , Animals , Arginine , Cancellous Bone/growth & development , Cortical Bone/growth & development , Female , Fibroblasts/metabolism , Galectin 3/genetics , Humans , Male , Mice , Mice, Knockout , Mutation , Polysaccharides , Serine/genetics
2.
Front Endocrinol (Lausanne) ; 13: 910901, 2022.
Article in English | MEDLINE | ID: mdl-36046782

ABSTRACT

Unloading associated with spaceflight results in bone loss and increased fracture risk. Bone morphogenetic protein 2 (BMP2) is known to enhance bone formation, in part, through molecular pathways associated with mechanical loading; however, the effects of BMP2 during spaceflight remain unclear. Here, we investigated the systemic effects of BMP2 on mice sustaining a femoral fracture followed by housing in spaceflight (International Space Station or ISS) or on Earth. We hypothesized that in spaceflight, the systemic effects of BMP2 on weight-bearing bones would be blunted compared to that observed on Earth. Nine-week-old male mice were divided into four groups: 1) Saline+Earth; 2) BMP+Earth; 3) Saline+ISS; and 4) BMP+ISS (n = 10 mice/group, but only n = 5 mice/group were reserved for micro-computed tomography analyses). All mice underwent femoral defect surgery and were followed for approximately 4 weeks. We found a significant reduction in trabecular separation within the lumbar vertebrae after administering BMP2 at the fracture site of mice housed on Earth. In contrast, BMP2 treatment led to a significant increase in trabecular separation concomitant with a reduction in trabecular number within spaceflown tibiae. Although these and other lines of evidence support our hypothesis, the small sample size associated with rodent spaceflight studies limits interpretations. That said, it appears that a locally applied single dose of BMP2 at the femoral fracture site can have a systemic impact on distant bones, affecting bone quantity in several skeletal sites. Moreover, our results suggest that BMP2 treatment works through a pathway involving mechanical loading in which the best outcomes during its treatment on Earth occurred in the weight-bearing bones and in spaceflight occurred in bones subjected to higher muscle contraction.


Subject(s)
Femoral Fractures , Space Flight , Animals , Bone Morphogenetic Protein 2 , Bone and Bones , Femoral Fractures/diagnostic imaging , Femoral Fractures/etiology , Male , Mice , X-Ray Microtomography
3.
NPJ Microgravity ; 7(1): 12, 2021 Mar 26.
Article in English | MEDLINE | ID: mdl-33772025

ABSTRACT

With increased human presence in space, bone loss and fractures will occur. Thrombopoietin (TPO) is a recently patented bone healing agent. Here, we investigated the systemic effects of TPO on mice subjected to spaceflight and sustaining a bone fracture. Forty, 9-week-old, male, C57BL/6 J were divided into 4 groups: (1) Saline+Earth; (2) TPO + Earth; (3) Saline+Flight; and (4) TPO + Flight (n = 10/group). Saline- and TPO-treated mice underwent a femoral defect surgery, and 20 mice were housed in space ("Flight") and 20 mice on Earth for approximately 4 weeks. With the exception of the calvarium and incisor, positive changes were observed in TPO-treated, spaceflight bones, suggesting TPO may improve osteogenesis in the absence of mechanical loading. Thus, TPO, may serve as a new bone healing agent, and may also improve some skeletal properties of astronauts, which might be extrapolated for patients on Earth with restraint mobilization and/or are incapable of bearing weight on their bones.

4.
Bone ; 146: 115883, 2021 05.
Article in English | MEDLINE | ID: mdl-33581374

ABSTRACT

Angiogenesis is a vital process during the regeneration of bone tissue. The aim of this study was to investigate angiogenesis at the fracture site as well as at distal locations from obesity-induced type 2 diabetic mice that were treated with bone morphogenetic protein-2 (BMP-2, local administration at the time of surgery) to heal a femoral critical sized defect (CSD) or saline as a control. Mice were fed a high fat diet (HFD) to induce a type 2 diabetic-like phenotype while low fat diet (LFD) animals served as controls. Endothelial cells (ECs) were isolated from the lungs (LECs) and bone marrow (BMECs) 3 weeks post-surgery, and the fractured femurs were also examined. Our studies demonstrate that local administration of BMP-2 at the fracture site in a CSD model results in complete bone healing within 3 weeks for all HFD mice and 66.7% of LFD mice, whereas those treated with saline remain unhealed. At the fracture site, vessel parameters and adipocyte numbers were significantly increased in BMP-2 treated femurs, irrespective of diet. At distal sites, LEC and BMEC proliferation was not altered by diet or BMP-2 treatment. HFD increased the tube formation ability of both LECs and BMECs. Interestingly, BMP-2 treatment at the time of surgery reduced tube formation in LECs and humeri BMECs. However, migration of BMECs from HFD mice treated with BMP-2 was increased compared to BMECs from HFD mice treated with saline. BMP-2 treatment significantly increased the expression of CD31, FLT-1, and ANGPT2 in LECs and BMECs in LFD mice, but reduced the expression of these same genes in HFD mice. To date, this is the first study that depicts the systemic influence of fracture surgery and local BMP-2 treatment on the proliferation and angiogenic potential of ECs derived from the bone marrow and lungs.


Subject(s)
Diabetes Mellitus, Experimental , Femoral Fractures , Animals , Cell Proliferation , Diet, High-Fat/adverse effects , Endothelial Cells , Mice
5.
Aging (Albany NY) ; 12(14): 15121-15133, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32634116

ABSTRACT

Megakaryocytes (MKs) support bone formation by stimulating osteoblasts (OBs) and inhibiting osteoclasts (OCs). Aging results in higher bone resorption, leading to bone loss. Whereas previous studies showed the effects of aging on MK-mediated bone formation, the effects of aging on MK-mediated OC formation is poorly understood. Here we examined the effect of thrombopoietin (TPO) and MK-derived conditioned media (CM) from young (3-4 months) and aged (22-25 months) mice on OC precursors. Our findings showed that aging significantly increased OC formation in vitro. Moreover, the expression of the TPO receptor, Mpl, and circulating TPO levels were elevated in the bone marrow cavity. We previously showed that MKs from young mice secrete factors that inhibit OC differentiation. However, rather than inhibiting OC development, we found that MKs from aged mice promote OC formation. Interestingly, these age-related changes in MK functionality were only observed using female MKs, potentially implicating the sex steroid, estrogen, in signaling. Further, RANKL expression was highly elevated in aged MKs suggesting MK-derived RANKL signaling may promote osteoclastogenesis in aging. Taken together, these data suggest that modulation in TPO-Mpl expression in bone marrow and age-related changes in the MK secretome promote osteoclastogenesis to impact skeletal aging.


Subject(s)
Aging/physiology , Bone Marrow , Bone Resorption/metabolism , Megakaryocytes/physiology , Osteogenesis/physiology , RANK Ligand/metabolism , Receptors, Thrombopoietin/metabolism , Thrombopoietin/metabolism , Age Factors , Animals , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Differentiation , Cell Proliferation , Estrogens/metabolism , Mice , Sex Factors , Signal Transduction/physiology
6.
Calcif Tissue Int ; 106(3): 283-293, 2020 03.
Article in English | MEDLINE | ID: mdl-31745588

ABSTRACT

Sex hormone deprivation commonly occurs following menopause in women or after androgen-depletion during prostate cancer therapy in men, resulting in rapid bone turnover and loss of bone mass. There is a need to identify novel therapies to improve bone mass in these conditions. Previously, we identified age- and sex-dependent effects on bone mass in mice with deletion of the gene encoding the ß-galactoside binding lectin, galectin-3 (Lgals3-KO). Due to the influence of sex on the phenotype, we tested the role of sex hormones, estrogen (ß-estradiol; E2), and androgen (5α-dihydroxytestosterone; DHT) in Lgals3-KO mice. To address this, we subjected male and female wild-type and Lgals3-KO mice to gonadectomy ± E2 or DHT rescue and compared differential responses in bone mass and bone formation. Following gonadectomy, male and female Lgals3-KO mice had greater cortical bone expansion (increased total area; T.Ar) and reduced loss of bone area (B.Ar). While T.Ar and B.Ar were increased in response to DHT in wild-type mice, DHT did not alter these parameters in Lgals3-KO mice. E2 rescue more strongly increased B.Ar in Lgals3-KO compared to wild-type female mice due to a failure of E2 to repress the increase in T.Ar following gonadectomy. Lgals3-KO mice had more osteoblasts relative to bone surface when compared to wild-type animals in sham, gonadectomy, and E2 rescue groups. DHT suppressed this increase. This study revealed a mechanism for the sex-dependency of the Lgals3-KO aging bone phenotype and supports targeting galectin-3 to protect against bone loss associated with decreased sex hormone production.


Subject(s)
Cortical Bone , Galectin 3/genetics , Osteoporosis/etiology , Androgens/pharmacology , Animals , Body Composition , Cancellous Bone/diagnostic imaging , Cancellous Bone/drug effects , Castration , Cortical Bone/diagnostic imaging , Cortical Bone/drug effects , Dihydrotestosterone/pharmacology , Estrogens/pharmacology , Female , Male , Mice , Mice, Knockout , Osteoporosis/diagnostic imaging , Osteoporosis/genetics
7.
NPJ Microgravity ; 5: 21, 2019.
Article in English | MEDLINE | ID: mdl-31583271

ABSTRACT

Gravity has an important role in both the development and maintenance of bone mass. This is most evident in the rapid and intense bone loss observed in both humans and animals exposed to extended periods of microgravity in spaceflight. Here, cohabitating 9-week-old male C57BL/6 mice resided in spaceflight for ~4 weeks. A skeletal survey of these mice was compared to both habitat matched ground controls to determine the effects of microgravity and baseline samples in order to determine the effects of skeletal maturation on the resulting phenotype. We hypothesized that weight-bearing bones would experience an accelerated loss of bone mass compared to non-weight-bearing bones, and that spaceflight would also inhibit skeletal maturation in male mice. As expected, spaceflight had major negative effects on trabecular bone mass of the following weight-bearing bones: femur, tibia, and vertebrae. Interestingly, as opposed to the bone loss traditionally characterized for most weight-bearing skeletal compartments, the effects of spaceflight on the ribs and sternum resembled a failure to accumulate bone mass. Our study further adds to the insight that gravity has site-specific influences on the skeleton.

8.
Sci Rep ; 9(1): 11419, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31388031

ABSTRACT

Spaceflight results in reduced mechanical loading of the skeleton, which leads to dramatic bone loss. Low bone mass is associated with increased fracture risk, and this combination may compromise future, long-term, spaceflight missions. Here, we examined the systemic effects of spaceflight and fracture surgery/healing on several non-injured bones within the axial and appendicular skeleton. Forty C57BL/6, male mice were randomized into the following groups: (1) Sham surgery mice housed on the earth (Ground + Sham); (2) Femoral segmental bone defect surgery mice housed on the earth (Ground + Surgery); (3) Sham surgery mice housed in spaceflight (Flight + Sham); and (4) Femoral segmental bone defect surgery mice housed in spaceflight (Flight + Surgery). Mice were 9 weeks old at the time of launch and were euthanized approximately 4 weeks after launch. Micro-computed tomography (µCT) was used to evaluate standard bone parameters in the tibia, humerus, sternebra, vertebrae, ribs, calvarium, mandible, and incisor. One intriguing finding was that both spaceflight and surgery resulted in virtually identical losses in tibial trabecular bone volume fraction, BV/TV (24-28% reduction). Another important finding was that surgery markedly changed tibial cortical bone geometry. Understanding how spaceflight, surgery, and their combination impact non-injured bones will improve treatment strategies for astronauts and terrestrial humans alike.


Subject(s)
Bone Density/physiology , Fracture Healing/physiology , Fractures, Bone/surgery , Space Flight , Animals , Disease Models, Animal , Female , Femur/diagnostic imaging , Femur/physiology , Fractures, Bone/diagnostic imaging , Fractures, Bone/physiopathology , Humans , Male , Mandible/diagnostic imaging , Mandible/physiology , Mice , Tibia/diagnostic imaging , Tibia/physiology , Weight-Bearing/physiology , X-Ray Microtomography
9.
Bone ; 127: 452-459, 2019 10.
Article in English | MEDLINE | ID: mdl-31299382

ABSTRACT

Osteoblast number and activity decreases with aging, contributing to the age-associated decline of bone mass, but the mechanisms underlying changes in osteoblast activity are not well understood. Here, we show that the age-associated bone loss critically depends on impairment of the ability of megakaryocytes (MKs) to support osteoblast proliferation. Co-culture of osteoblast precursors with young MKs is known to increase osteoblast proliferation and bone formation. However, co-culture of osteoblast precursors with aged MKs resulted in significantly fewer osteoblasts compared to co-culture with young MKs, and this was associated with the downregulation of transforming growth factor beta. In addition, the ability of MKs to increase bone mass was attenuated during aging as transplantation of GATA1low/low hematopoietic donor cells (which have elevated MKs/MK precursors) from young mice resulted in an increase in bone mass of recipient mice compared to transplantation of young wild-type donor cells, whereas transplantation of GATA1low/low donor cells from old mice failed to enhance bone mass in recipient mice compared to transplantation of old wild-type donor cells. These findings suggest that the preservation or restoration of the MK-mediated induction of osteoblast proliferation during aging may hold the potential to prevent age-associated bone loss and resulting fractures.


Subject(s)
Aging/physiology , Bone and Bones/anatomy & histology , Megakaryocytes/cytology , Osteoblasts/cytology , Adoptive Transfer , Animals , Bone Marrow/metabolism , Bone and Bones/diagnostic imaging , Cell Count , Cell Proliferation , GATA1 Transcription Factor/metabolism , Hematopoietic Stem Cells/metabolism , Male , Mice, Inbred C57BL , Organ Size , Phenotype , X-Ray Microtomography
10.
Stem Cells Dev ; 27(10): 671-682, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29631496

ABSTRACT

Emerging evidence demonstrates that megakaryocytes (MK) play key roles in regulating skeletal homeostasis and hematopoiesis. To test if the loss of MK negatively impacts osteoblastogenesis and hematopoiesis, we generated conditional knockout mice where Mpl, the receptor for the main MK growth factor, thrombopoietin, was deleted specifically in MK (Mplf/f;PF4cre). Unexpectedly, at 12 weeks of age, these mice exhibited a 10-fold increase in platelets, a significant expansion of hematopoietic/mesenchymal precursors, and a remarkable 20-fold increase in femoral midshaft bone volume. We then investigated whether MK support hematopoietic stem cell (HSC) function through the interaction of MK with osteoblasts (OB). LSK cells (Lin-Sca1+CD117+, enriched HSC population) were co-cultured with OB+MK for 1 week (1wk OB+MK+LSK) or OB alone (1wk OB+LSK). A significant increase in colony-forming units was observed with cells from 1wk OB+MK cultures. Competitive repopulation studies demonstrated significantly higher engraftment in mice transplanted with cells from 1wk OB+MK+LSK cultures compared to 1wk OB+LSK or LSK cultured alone for 1 week. Furthermore, single-cell expression analysis of OB cultured±MK revealed adiponectin as the most significantly upregulated MK-induced gene, which is required for optimal long-term hematopoietic reconstitution. Understanding the interactions between MK, OB, and HSC can inform the development of novel treatments to enhance both HSC recovery following myelosuppressive injuries, as well as bone loss diseases, such as osteoporosis.


Subject(s)
Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Megakaryocytes/cytology , Osteoblasts/cytology , Animals , Cells, Cultured , Coculture Techniques/methods , Female , Hematopoietic Stem Cells/metabolism , Male , Megakaryocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoblasts/metabolism , Thrombopoietin/metabolism
11.
Bone Res ; 6: 7, 2018.
Article in English | MEDLINE | ID: mdl-30886760

ABSTRACT

Imbalances between bone formation and bone resorption, which can occur due to aging or sex hormone deprivation, result in decreased bone mass and an increased risk of fracture. Previous studies have suggested that the ß-galactoside binding lectin, galectin-3, is involved in bone remodeling. We compared bone parameters of mice having null alleles of the galectin-3 gene (Lgals3-KO) with those of their wild-type littermates. Lgals3 deficiency increased cortical bone expansion at 36 weeks (wk) and preserved or enhanced bone mass in both male and female mutant mice. In addition, female Lgals3-KO mice were protected from age-related loss of trabecular bone. Histomorphometry and ex vivo primary cell differentiation assays showed increased osteoblastogenesis with little-to-no effect on osteoclastogenesis, suggesting the increased bone mass phenotype is primarily due to increased anabolism. Our study identifies galectin-3 as a negative regulator of bone formation and suggests that disruption of galectin-3 may be useful in preventing bone loss during aging.

12.
J Bone Miner Res ; 32(12): 2405-2414, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28777473

ABSTRACT

In academia, authorship is considered a currency and is important for career advancement. As the Journal of Bone and Mineral Research (JBMR) is the highest-ranked journal in the field of bone, muscle, and mineral metabolism and is the official publication of the American Society for Bone and Mineral Research, we sought to examine authorship changes over JBMR's 30-year history. Two bibliometric methods were used to collect the data. The "decade method" included all published manuscripts throughout 1 year in each decade over the past 30 years starting with the inaugural year, yielding 746 manuscripts for analysis. The "random method" examined 10% of published manuscripts from each of the 30 years, yielding 652 manuscripts for analysis. Using both methods, the average number of authors per manuscript, numerical location of the corresponding author, number of collaborating institutions, number of collaborating countries, number of printed manuscript pages, and the number of times each manuscript was cited all significantly increased between 1986 and 2015 (p < 10-4 ). Using the decade method, there was a significant increase in the percentage of female first authors over time from 35.8% in 1986 to 47.7% in 2015 (p = 0.02), and this trend was confirmed using the random method. The highest percentage of female first authors in 2015 was in Europe (60.0%), and Europe also had the most dramatic increase in female first authors over time (more than double in 2015 compared with 1986). Likewise, the overall number of female corresponding authors significantly increased during the past 30 years. With the increasing demands of publishing in academic medicine, understanding changes in publishing characteristics over time and by geographical region is important. These findings highlight JBMR's authorship trends over the past 30 years and demonstrate those countries having the most changes and where challenges still exist. © 2017 American Society for Bone and Mineral Research.


Subject(s)
Authorship , Bibliometrics , Cooperative Behavior , Publications , Female , Geography , Humans , Male , Sex Characteristics , Time Factors
13.
Bone ; 98: 68-78, 2017 05.
Article in English | MEDLINE | ID: mdl-28384511

ABSTRACT

Inactivating mutations that lead to loss of heterozygosity within the HRPT2/Cdc73 gene are directly linked to the development of primary hyperparathyroidism, parathyroid adenomas, and ossifying fibromas of the jaw (HPT-JT). The protein product of the Cdc73 gene, parafibromin, is a core member of the polymerase-associated factors (PAF) complex, which coordinates epigenetic modifiers and transcriptional machinery to control gene expression. We conditionally deleted Cdc73 within mesenchymal progenitors or within mature osteoblasts and osteocytes to determine the consequences of parafibromin loss within the mesenchymal lineage. Homozygous deletion of Cdc73 via the Dermo1-Cre driver resulted in embryos which lacked mesenchymal organ development of internal organs, including the heart and fetal liver. Immunohistochemical detection of cleaved caspase-3 revealed extensive apoptosis within the progenitor pools of developing organs. Unexpectedly, when Cdc73 was homozygously deleted within mature osteoblasts and osteocytes (via the Ocn-Cre driver), the mice had a normal life span but increased cortical and trabecular bone. OCN-Cre;Cdc73flox/flox bones displayed large cortical pores actively undergoing bone remodeling. Additionally the cortical bone of OCN-Cre;Cdc73flox/flox femurs contained osteocytes with marked amounts of cytoplasmic RNA and a high rate of apoptosis. Transcriptional analysis via RNA-seq within OCN-Cre;Cdc73flox/flox osteoblasts showed that loss of Cdc73 led to a derepression of osteoblast-specific genes, specifically those for collagen and other bone matrix proteins. These results aid in our understanding of the role parafibromin plays within transcriptional regulation, terminal differentiation, and bone homeostasis.


Subject(s)
Bone Remodeling/physiology , Bone and Bones/metabolism , Osteoblasts/metabolism , Tumor Suppressor Proteins/metabolism , Absorptiometry, Photon , Animals , Cell Differentiation/physiology , Flow Cytometry , Immunohistochemistry , In Situ Nick-End Labeling , Mice , Mice, Knockout , Mice, Mutant Strains , Osteogenesis , Transcriptome , X-Ray Microtomography
14.
Methods Mol Biol ; 1273: 203-14, 2015.
Article in English | MEDLINE | ID: mdl-25753713

ABSTRACT

The glycan array is a powerful tool for investigating the specificities of glycan-binding proteins. By incubating a glycan-binding protein on a glycan array, the relative binding to hundreds of different oligosaccharides can be quantified in parallel. Based on these data, much information can be obtained about the preference of a glycan-binding protein for specific subcomponents of oligosaccharides or motifs. In many cases, the analysis and interpretation of glycan array data can be time consuming and imprecise if done manually. Recently we developed software, called GlycoSearch, to facilitate the analysis and interpretation of glycan array data based on the previously developed methods called Motif Segregation and Outlier Motif Analysis. Here we describe the principles behind the software, the use of the software, and an example application. The automated, objective, and precise analysis of glycan array data should enhance the value of the data for a broad range of research applications.


Subject(s)
Computational Biology/methods , Glycomics/methods , Polysaccharides/metabolism , Software , Statistics as Topic , Plant Lectins/chemistry , Protein Binding
15.
BMC Biotechnol ; 14: 101, 2014 Dec 06.
Article in English | MEDLINE | ID: mdl-25479762

ABSTRACT

BACKGROUND: γ-Glutamyl transpeptidase 1 (GGT1) is an N-glycosylated membrane protein that catabolizes extracellular glutathione and other γ-glutamyl-containing substrates. In a variety of disease states, including tumor formation, the enzyme is shed from the surface of the cell and can be detected in serum. The structures of the N-glycans on human GGT1 (hGGT1) have been shown to be tissue-specific. Tumor-specific changes in the glycans have also been observed, suggesting that the N-glycans on hGGT1 would be an important biomarker for detecting tumors and monitoring their progression during treatment. However, the large quantities of purified protein required to fully characterize the carbohydrate content poses a significant challenge for biomarker development. Herein, we investigated a new antibody-lectin sandwich array (ALSA) platform to determine whether this microanalytical technique could be applied to the characterization of N-glycan content of hGGT1 in complex biological samples. RESULTS: Our data show that hGGT1 can be isolated from detergent extracted membrane proteins by binding to the ALSA platform. Probing hGGT1 with lectins enables characterization of the N-glycans. We probed hGGT1 from normal human liver tissue, normal human kidney tissue, and hGGT1 expressed in the yeast Pichia pastoris. The lectin binding patterns obtained with the ALSA platform are consistent with the hGGT1 N-glycan composition obtained from previous large-scale hGGT1 N-glycan characterizations from these sources. We also validate the implementation of the Microcystis aeruginosa lectin, microvirin, in this platform and provide refined evidence for its efficacy in specifically recognizing high-mannose-type N-glycans, a class of carbohydrate modification that is distinctive of hGGT1 expressed by many tumors. CONCLUSION: Using this microanalytical approach, we provide proof-of-concept for the implementation of ALSA in conducting high-throughput studies aimed at investigating disease-related changes in the glycosylation patterns on hGGT1 with the goal of enhancing clinical diagnoses and targeted treatment regimens.


Subject(s)
Protein Array Analysis/methods , gamma-Glutamyltransferase/metabolism , Antibodies/chemistry , Glycosylation , Humans , Kidney/chemistry , Kidney/enzymology , Lectins/chemistry , Liver/chemistry , Liver/enzymology , Protein Binding , gamma-Glutamyltransferase/chemistry
16.
Immunol Cell Biol ; 91(7): 477-85, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23856766

ABSTRACT

Schistosome parasites follow a complex migration path through various tissues, changing their antigenic profile as they develop. A thorough understanding of the antibody response in each tissue region could help unravel the complex immunology of these developing parasites and aid vaccine design. Here we used a novel strategy for analysing the local antibody responses induced by Schistosoma japonicum infection at each site of infection. Cells from rat lymph nodes draining the sites of larval migration (the skin and lungs), the liver-lymph nodes where adults reside and the spleens were cultured to allow the in vivo-induced antibody-secreting cells to release antibody into the media. The amount and isotype of antibodies secreted in the supernatants differed significantly in the different lymph nodes and spleen, corresponding with the migration path of the schistosome worms. In addition, there were significant differences in binding specificity, as determined by surface labelling, western blots and by screening a glycan array. Through capturing the local antibody response, this study has revealed dramatic differences in the quality and specificity of the immune response at different tissue sites, and highlighted the existence of stage-specific protein and carbohydrate antigens. This will provide a valuable tool for the isolation of novel vaccine targets against the larval stages of schistosomes.


Subject(s)
Antibodies, Helminth/metabolism , Lymph Nodes/metabolism , Organ Specificity , Schistosoma japonicum/physiology , Schistosomiasis japonica/immunology , Animals , Antigens, Helminth/immunology , Cells, Cultured , Epitopes/immunology , Female , Humans , Immunity, Humoral , Larva , Liver/immunology , Liver/parasitology , Lung/immunology , Lung/parasitology , Lymph Nodes/immunology , Organ Specificity/immunology , Rats , Rats, Wistar , Skin/immunology , Skin/parasitology
17.
PLoS One ; 8(6): e65905, 2013.
Article in English | MEDLINE | ID: mdl-23750276

ABSTRACT

The development of accurate clinical biomarkers has been challenging in part due to the diversity between patients and diseases. One approach to account for the diversity is to use multiple markers to classify patients, based on the concept that each individual marker contributes information from its respective subclass of patients. Here we present a new strategy for developing biomarker panels that accounts for completely distinct patient subclasses. Marker State Space (MSS) defines "marker states" based on all possible patterns of high and low values among a panel of markers. Each marker state is defined as either a case state or a control state, and a sample is classified as case or control based on the state it occupies. MSS was used to define multi-marker panels that were robust in cross validation and training-set/test-set analyses and that yielded similar classification accuracy to several other classification algorithms. A three-marker panel for discriminating pancreatic cancer patients from control subjects revealed subclasses of patients based on distinct marker states. MSS provides a straightforward approach for modeling highly divergent subclasses of patients, which may be adaptable for diverse applications.


Subject(s)
Computational Biology/methods , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/diagnosis , Biomarkers/blood , Diagnosis, Differential , Humans , Reproducibility of Results , Software
18.
Bone Res ; 1(1): 27-71, 2013 Mar.
Article in English | MEDLINE | ID: mdl-26273492

ABSTRACT

The Wnt signaling pathway plays key roles in differentiation and development and alterations in this signaling pathway are causally associated with numerous human diseases. While several laboratories were examining roles for Wnt signaling in skeletal development during the 1990s, interest in the pathway rose exponentially when three key papers were published in 2001-2002. One report found that loss of the Wnt co-receptor, Low-density lipoprotein related protein-5 (LRP5), was the underlying genetic cause of the syndrome Osteoporosis pseudoglioma (OPPG). OPPG is characterized by early-onset osteoporosis causing increased susceptibility to debilitating fractures. Shortly thereafter, two groups reported that individuals carrying a specific point mutation in LRP5 (G171V) develop high-bone mass. Subsequent to this, the causative mechanisms for these observations heightened the need to understand the mechanisms by which Wnt signaling controlled bone development and homeostasis and encouraged significant investment from biotechnology and pharmaceutical companies to develop methods to activate Wnt signaling to increase bone mass to treat osteoporosis and other bone disease. In this review, we will briefly summarize the cellular mechanisms underlying Wnt signaling and discuss the observations related to OPPG and the high-bone mass disorders that heightened the appreciation of the role of Wnt signaling in normal bone development and homeostasis. We will then present a comprehensive overview of the core components of the pathway with an emphasis on the phenotypes associated with mice carrying genetically engineered mutations in these genes and clinical observations that further link alterations in the pathway to changes in human bone.

19.
Proteomics ; 12(13): 2212-20, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22623153

ABSTRACT

The CA 19-9 antigen is currently the best individual marker for the detection of pancreatic cancer. In order to optimize the CA 19-9 assay and to develop approaches to further improve cancer detection, it is important to understand the specificity differences between CA 19-9 antibodies and the consequential affect on biomarker performance. Antibody arrays enabled multiplexed comparisons between five different CA 19-9 antibodies used in the analysis of plasma samples from pancreatic cancer patients and controls. Major differences were observed between antibodies in their detection of particular patient samples. Glycan array analysis revealed that certain antibodies were highly specific for the canonical CA 19-9 epitope, sialyl-Lewis A, while others bound sialyl-Lewis A in addition to a related structure called sialyl-Lewis C and modification with Nue5Gc. In a much larger patient cohort, we confirmed the binding of sialyl-Lewis C glycan by one of the antibodies and showed that the broader specificity led to the detection of an increased number of cancer patients without increasing detection of pancreatitis patient samples. This work demonstrates that variation between antibody specificity for cancer-associated glycans can have significant implications for biomarker performance and highlights the value of characterizing and detecting the range of glycan structures that are elevated in cancer.


Subject(s)
Antibodies, Monoclonal/immunology , Biomarkers, Tumor/immunology , CA-19-9 Antigen/immunology , Pancreatic Neoplasms/immunology , Antibody Specificity , Biomarkers, Tumor/blood , CA-19-9 Antigen/blood , Carbohydrate Sequence , Cohort Studies , Humans , Molecular Sequence Data , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/diagnosis , Polysaccharides/chemistry , Polysaccharides/immunology , Protein Array Analysis
20.
J Proteome Res ; 11(5): 2904-11, 2012 May 04.
Article in English | MEDLINE | ID: mdl-22439797

ABSTRACT

Significant efforts are underway to develop new biomarkers from pancreatic cyst fluid. Previous research has made use of cyst fluid collected from surgically removed cysts, but the clinical implementation of biomarkers would use cyst fluid collected by endoscopic ultrasound-guided, fine-needle aspiration (EUS-FNA). The purpose of this study was to investigate the clinical applicability of cyst fluid research obtained using surgical specimens. Matched pairs of operating-room collected (OR) and EUS-FNA samples from 12 patients were evaluated for the levels of three previously described biomarkers, CA 19-9, CEA, and glycan levels detected by wheat germ agglutinin on MUC5AC (MUC5AC-WGA). CA 19-9 and MUC5AC-WGA correlated well between the sample types, although CEA was more variable between the sample types for certain patients. The variability was not due to the time delay between EUS-FNA and OR collection or differences in total protein concentrations but may be caused by contamination of the cyst fluid with blood proteins. The classification of each patient based on thresholds for each marker was perfectly consistent between sample types for CA 19-9 and MUC5AC-WGA and mostly consistent for CEA. Therefore, results obtained using OR-collected pancreatic cyst fluid samples should reliably transfer to the clinical setting using EUS-FNA samples.


Subject(s)
Biopsy, Fine-Needle/methods , Cyst Fluid/chemistry , Endosonography/methods , Pancreatic Cyst/chemistry , Biomarkers/chemistry , Blood Proteins/metabolism , CA-19-9 Antigen/chemistry , Humans , Mucin 5AC/chemistry , Operating Rooms/methods , Pancreatic Cyst/pathology , Pancreatic Cyst/surgery , Reproducibility of Results , Specimen Handling
SELECTION OF CITATIONS
SEARCH DETAIL
...