Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Nat Commun ; 12(1): 4475, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34294719

ABSTRACT

High Arctic ecosystems and Indigenous livelihoods are tightly linked and exposed to climate change, yet assessing their sensitivity requires a long-term perspective. Here, we assess the vulnerability of the North Water polynya, a unique seaice ecosystem that sustains the world's northernmost Inuit communities and several keystone Arctic species. We reconstruct mid-to-late Holocene changes in sea ice, marine primary production, and little auk colony dynamics through multi-proxy analysis of marine and lake sediment cores. Our results suggest a productive ecosystem by 4400-4200 cal yrs b2k coincident with the arrival of the first humans in Greenland. Climate forcing during the late Holocene, leading to periods of polynya instability and marine productivity decline, is strikingly coeval with the human abandonment of Greenland from c. 2200-1200 cal yrs b2k. Our long-term perspective highlights the future decline of the North Water ecosystem, due to climate warming and changing sea-ice conditions, as an important climate change risk.

2.
PLoS One ; 9(12): e114686, 2014.
Article in English | MEDLINE | ID: mdl-25531770

ABSTRACT

Hyperkalemia, a condition in which serum potassium ions (K+) exceed 5.0 mmol/L, is a common electrolyte disorder associated with substantial morbidity. Current methods of managing hyperkalemia, including organic polymer resins such as sodium polystyrene sulfonate (SPS), are poorly tolerated and/or not effective. Sodium zirconium cyclosilicate (ZS-9) is under clinical development as an orally administered, non-absorbed, novel, inorganic microporous zirconium silicate compound that selectively removes excess K+ in vivo. The development, structure and ion exchange properties of ZS-9 and its hypothesized mechanism of action are described. Based on calculation of the interatomic distances between the atoms forming the ZS-9 micropores, the size of the pore opening was determined to be ∼ 3 Š(∼ diameter of unhydrated K+). Unlike nonspecific organic polymer resins like SPS, the ZS-9 K+ exchange capacity (KEC) was unaffected by the presence of calcium (Ca2+) or magnesium ions (Mg2+) and showed>25-fold selectivity for K+ over either Ca2+ or Mg2+. Conversely, the selectivity of SPS for K+ was only 0.2-0.3 times its selectivity for Ca2+ or Mg2+in mixed ionic media. It is hypothesized that the high K+ specificity of ZS-9 is attributable to the chemical composition and diameter of the micropores, which possibly act in an analogous manner to the selectivity filter utilized by physiologic K+ channels. This hypothesized mechanism of action is supported by the multi-ion exchange studies. The effect of pH on the KEC of ZS-9 was tested in different media buffered to mimic different portions of the human gastrointestinal tract. Rapid K+ uptake was observed within 5 minutes - mainly in the simulated small intestinal and large intestinal fluids, an effect that was sustained for up to 1 hour. If approved, ZS-9 will represent a novel, first-in-class therapy for hyperkalemia with improved capacity, selectivity, and speed for entrapping K+ when compared to currently available options.


Subject(s)
Potassium/chemistry , Silicates/chemistry , Calcium/chemistry , Humans , Hydrogen-Ion Concentration , Hyperkalemia/drug therapy , Hyperkalemia/metabolism , Hyperkalemia/pathology , Ion Exchange , Ions/chemistry , Porosity , Potassium/blood , Silicates/therapeutic use , Sodium/chemistry
3.
Pigment Cell Melanoma Res ; 27(3): 408-17, 2014 May.
Article in English | MEDLINE | ID: mdl-24406016

ABSTRACT

Using the Xiphophorus fish melanoma model, we show a strong male bias for sunlight-induced malignant melanoma, consistent with that seen in the human population. To examine underlying factors, we exposed adult X. couchianus fish to a single, sublethal dose of UVB and measured circulating sex steroid hormones and expression of associated hormone receptor genes over a 24-h period. We found that a single exposure had profound effects on circulating levels of steroid hormones with significant decreases for all free sex steroids at 6 and 24 h and increases in conjugated 2-estradiol and 11-ketotestosterone at 6 and 24 h, respectively. Whereas ARα expression increased in male and female skin, neither ARß nor either of the ERs showed significant responses to UVB in either sex. The rapid response of male androgens and their receptors in the skin after UVB irradiation implicates hormones in the male bias of skin cancer and suggests that the photoendocrine response immediately after UV exposure may be relevant to melanomagenesis.


Subject(s)
Cyprinodontiformes/genetics , Gonadal Steroid Hormones/biosynthesis , Melanoma, Experimental/genetics , Models, Animal , Neoplasms, Radiation-Induced/genetics , Receptors, Androgen/biosynthesis , Receptors, Estrogen/biosynthesis , Skin/radiation effects , Ultraviolet Rays/adverse effects , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/radiation effects , DNA Damage , Female , Fish Diseases/epidemiology , Gonadal Steroid Hormones/genetics , Humans , Hydrocortisone/biosynthesis , Incidence , Male , Melanoma/epidemiology , Melanoma/veterinary , Melanoma, Experimental/etiology , Neoplasms, Radiation-Induced/etiology , Oxidative Stress , Receptors, Androgen/genetics , Receptors, Estrogen/genetics , Sex Distribution , Skin/metabolism
4.
Calcif Tissue Int ; 94(1): 78-87, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24101233

ABSTRACT

Throughout life, a balance exists within the marrow cavity between adipose tissue and bone. Each tissue derives from a common progenitor cell known both as a "bone marrow-derived multipotent stromal cell" and as a "mesenchymal stem cell" (BMSC). The majority of in vitro and in vivo data suggest that BMSCs differentiate into adipocytes or osteoblasts in a reciprocal manner. For example, while ligand induction of the transcription factors peroxisome proliferator-activated receptor γ initiates BMSC adipogenesis, it suppresses osteogenesis. Nevertheless, this hypothesis may oversimplify a complex regulatory paradigm. The picture may be further complicated by the systemic impact of extramedullary adipose depots on bone via the secretion of protein adipokines and lipid metabolites. This review focuses on past and current literature examining the mechanisms governing the adipose-bone interface.


Subject(s)
Adipocytes/metabolism , Bone Remodeling/physiology , Adipocytes/cytology , Animals , Humans , Mesenchymal Stem Cells/cytology , Osteoblasts/metabolism , Osteogenesis/physiology , Transcription Factors/metabolism
5.
Clin Biochem ; 45(12): 874-9, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22429519

ABSTRACT

OBJECTIVES: The authors have set out to evaluate the literature relevant to the dynamic regulation of adipogenesis and osteogenesis. DESIGN AND METHODS: A detailed search of the past and recent literature was conducted on Pubmed using a combination of keywords including: adipogenesis, bone marrow, hematopoiesis, mesenchymal stromal/stem cell, and osteogenesis. RESULTS: Throughout one's lifespan, the bone marrow microenvironment provides a unique niche for mesenchymal stromal/stem cells (BMSCs) and hematopoietic stem cells (HSCs). The marrow changes as a function of biological age and pathophysiology. Historically, clinical biochemistry has observed these changes from an HSC and hematological perspective. Nevertheless, these changes also reflect the balance between BMSC adipogenic and osteogenic processes which can display an inverse or reciprocal relationship. Multiple hormonal factors and nuclear hormone receptor ligands and drugs are responsible for BMSC lineage selection. Data from a number of laboratories now implicates endocrine feedback loops between extramedullary adipose depots and the central nervous system. CONCLUSIONS: This concise review provides a perspective on the mechanisms regulating BMSC differentiation in the context of biological aging, obesity, and osteoporosis.


Subject(s)
Adipose Tissue, White/metabolism , Bone Marrow Cells/physiology , Bone and Bones/metabolism , Mesenchymal Stem Cells/physiology , Adipose Tissue, White/pathology , Aging/metabolism , Animals , Bone and Bones/pathology , Brain/metabolism , Brain/physiopathology , Cell Differentiation , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/physiology , Humans , Obesity/metabolism , Obesity/pathology , Osteoporosis/metabolism , Osteoporosis/pathology
6.
Ambio ; 41(1): 96-105, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22270709

ABSTRACT

The term "tipping point" has become increasingly popular in scientific and media usage to refer to the world in which we live, to describe the probable effects of the processes arising from the consequences of the ways we live with and transform the world, to describe our relationships with the environment, and the effects of our actions. Through this shift in scientific thinking, climate is talked about and represented as having the power to influence our lives in ways we have never before experienced or imagined, suggesting something transformative, disruptive, and decentering. Yet, in doing so, the complexity of the human world is explained in terms of scientific models that suggest a return to climatic determinism that simplifies our understanding of human-environment relations. How is anthropology to contribute to this discussion and to research and policy action on climate change? This article reflects on this and other questions as a way of contributing to the discussion of tipping points: are tipping points and thresholds metaphors, are they to be understood within contexts of speculative forecast, or are they descriptions of real events and indications of future change? What do we mean when we use these terms to imagine, describe, and represent the world? What relevance do they have for anticipatory knowledge and anticipatory practice? Indeed, how does anticipation guide us through a world of shifting conditions and sudden surprises and influence ways we orient ourselves toward the future?


Subject(s)
Climate Change , Climate , Environmental Monitoring/methods , Research Design/trends , Arctic Regions , Humans , Models, Theoretical , Policy
7.
Curr Pharm Des ; 17(4): 332-9, 2011.
Article in English | MEDLINE | ID: mdl-21375497

ABSTRACT

Information relating to the biology, culture expansion, and mechanisms relating to adipose-derived cells has advanced significantly in the past decade. Both the heterogeneous stromal vascular fraction (SVF) and more homogeneous adipose-derived stem cells (ASC) offer unique opportunities as novel cell-based therapeutics and as traditional pharmaceutical discovery tools. This review highlights the cytokine secretory functions of ASC and SVF cells as well as their potential use as immunomodulators and gene delivery vehicles. These functions make it feasible to exploit adipose-derived cells in the treatment of ischemic, musculoskeletal, and oncological disorders. With appropriate commercial development and in vivo validation, ASC and SVF cells will have a significant therapeutic impact in the future.


Subject(s)
Adipocytes/cytology , Adipocytes/metabolism , Adipose Tissue/metabolism , Adiposity , Drug Design , Regenerative Medicine/methods , Stem Cells/physiology , Stromal Cells/physiology , Adipose Tissue/blood supply , Adipose Tissue/cytology , Animals , Humans , Stromal Cells/cytology
8.
Transfus Med Hemother ; 35(3): 228-238, 2008.
Article in English | MEDLINE | ID: mdl-21547120

ABSTRACT

SUMMARY: Mesenchymal stem cells (MSCs) represent a class of multipotent progenitor cells that have been isolated from multiple tissue sites. Of these, adipose tissue and bone marrow offer advantages in terms of access, abundance, and the extent of their documentation in the literature. This review focuses on the in vitro differentiation capability of cells derived from adult human tissue. Multiple, independent studies have demonstrated that MSCs can commit to mesodermal (adipocyte, chondrocyte, hematopoietic support, myocyte, osteoblast, tenocyte), ectodermal (epithelial, glial, neural), and endodermal (hepatocyte, islet cell) lineages. The limitations and promises of these studies in the context of tissue engineering are discussed.

9.
Bone ; 39(6): 1361-72, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16904389

ABSTRACT

Whereas continuous PTH infusion increases bone resorption and bone loss, intermittent PTH treatment stimulates bone formation, in part, via reactivation of quiescent bone surfaces and reducing osteoblast apoptosis. We investigated the possibility that intermittent and continuous PTH treatment also differentially regulates osteogenic and adipocytic lineage commitment of bone marrow stromal progenitor/mesenchymal stem cells (MSC). The MSC were cultured under mildly adipogenic conditions in medium supplemented with dexamethasone, insulin, isobutyl-methylxanthine and troglitazone (DIIT), and treated with 50 nM human PTH(1-34) for either 1 h/day or continuously (PTH replenished every 48 h). After 6 days, cells treated with PTH for 1 h/day retained their normal fibroblastic appearance whereas those treated continuously adopted a polygonal, irregular morphology. After 12-18 days numerous lipid vacuole and oil red O-positive adipocytes had developed in cultures treated with DIIT alone, or with DIIT and continuous PTH. In contrast, adipocyte number was reduced and alkaline phosphatase staining increased in the cultures treated with DIIT and 1 h/day PTH, indicating suppression of adipogenesis and possible promotion of early osteoblastic differentiation. Furthermore, intermittent but not continuous PTH treatment suppressed markers of differentiated adipocytes such as mRNA expression of lipoprotein lipase and PPARgamma as well as glycerol 3-phosphate dehydrogenase activity. All of these effects of intermittent PTH were also produced by a 1 h/day treatment with AH3960 (30 microM), a small molecule, non-peptide agonist of the PTH1 receptor. AH3960, like PTH, activates both the cAMP and calcium signaling pathways. Treatment with the adenylyl cyclase activator forskolin for 1 h/day, mimicked the anti-adipogenic effect of intermittent PTH, whereas pretreatment with the protein kinase-A inhibitor H89 prior to intermittent PTH resulted in almost complete conversion to adipocytes. In contrast, the MAP kinase inhibitor PD 98059 failed to prevent the anti-adipocytic effect of intermittent PTH, suggesting that the inhibitory effect of PTH on adipocyte differentiation is predominantly cAMP-dependent. These results demonstrate a differential effect of PTH1 receptor agonists on the adipocytic commitment and differentiation of adult human bone marrow mesenchymal stem cells. This response may represent an additional mechanism that contributes to the overall bone anabolic action of intermittent PTH.


Subject(s)
Adipocytes/drug effects , Bone Marrow Cells/drug effects , Receptor, Parathyroid Hormone, Type 1/agonists , Teriparatide/pharmacology , Adenylyl Cyclases/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Alkaline Phosphatase/metabolism , Barbiturates/pharmacology , Base Sequence , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Cyclic AMP/metabolism , DNA Primers/genetics , Gene Expression/drug effects , Genetic Markers , Glycerolphosphate Dehydrogenase/metabolism , Humans , Lipid Metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Receptor, Parathyroid Hormone, Type 1/genetics , Stromal Cells/cytology , Stromal Cells/drug effects , Stromal Cells/metabolism
10.
J Cell Biochem ; 98(2): 251-66, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16479589

ABSTRACT

The relationship between bone and fat formation within the bone marrow microenvironment is complex and remains an area of active investigation. Classical in vitro and in vivo studies strongly support an inverse relationship between the commitment of bone marrow-derived mesenchymal stem cells or stromal cells to the adipocyte and osteoblast lineage pathways. In this review, we focus on the recent literature exploring the mechanisms underlying these differentiation events and discuss their implications relevant to osteoporosis and regenerative medicine.


Subject(s)
Adipose Tissue/cytology , Adipose Tissue/metabolism , Bone and Bones/metabolism , Mesenchymal Stem Cells/cytology , Adipocytes/cytology , Animals , Bone Density/physiology , Bone Diseases, Metabolic/physiopathology , Cell Differentiation , Child , Cholesterol/metabolism , Exercise/physiology , Hematopoiesis/physiology , Humans , Obesity/metabolism , Osteogenesis/physiology , Osteoporosis/physiopathology , Protease Inhibitors/pharmacology , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/metabolism , Signal Transduction/physiology
11.
PPAR Res ; 2006: 52950, 2006.
Article in English | MEDLINE | ID: mdl-17259665

ABSTRACT

PPARgamma plays a central role in the formation of fat. Regulation of PPARgamma activity depends on numerous factors ranging from dietary ligands to nuclear hormone coactivators and corepressors to oxygen-sensing mechanisms. In addition, the interplay of PPARgamma with other nuclear hormone receptors has implications for the balance between adipogenesis and osteogenesis in mesenchymal stem cells of the bone marrow stroma. This review will explore a range of factors influencing PPARgamma activity and how these interactions may affect osteogenesis.

12.
Bone ; 37(2): 159-69, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15921971

ABSTRACT

In this paper, we propose a mathematical model for parathyroid hormone receptor (PTH1R) kinetics, focusing on the receptor's response to PTH dosing to discern bone formation responses from bone resorption. The PTH1R is a major target for new osteoporosis treatments, as pulsatile PTH dosing has been shown to induce net bone formation in both animals and humans, and PTH(1-34) was recently FDA approved for the treatment of post-menopausal osteoporosis. PTH has also been shown to cause net bone loss when given continuously, so that the net action of PTH on bone is dependent on the dosing pattern. We have developed a simplified two-state receptor kinetics model for the PTH1R, based on the concepts of Segel et al., to distinguish the activity of active and inactive receptor and receptor-ligand complexes. The goal is to develop a plausible model of the minimal essential biological relationships necessary for understanding the responses to PTH dosing. A two-state model is able to effectively discriminate between continuous and pulsatile PTH dosing using the active species as surrogates for the downstream anabolic response. For continuous PTH dosing, the model predicts a desensitized system dominated by the inactive receptor and complex, consistent with downstream net bone loss that has been demonstrated experimentally. Using pulsatile PTH dosing, the model system predicts a highly sensitized state dominated by the active receptor and complex, corresponding to net bone formation. These results are consistent with the hypothesis that the kinetics of the receptor plays a critical role in the downstream effects of PTH dosing. Moreover, these results indicate that within a range of biologically relevant PTH doses, the two-state model is able to capture the differential behavior of the system for both continuous and pulsatile PTH dosing. The development of such a model provides a rational basis for developing more biologically extensive models that may support the design of optimal dosing strategies for PTH-based anti-osteoporosis treatments. Moreover, this model provides a unique starting point from which to design experiments investigating PTH receptor biology.


Subject(s)
Bone Resorption/therapy , Models, Biological , Parathyroid Hormone/administration & dosage , Receptor, Parathyroid Hormone, Type 1/physiology , Animals , Humans , Kinetics , Parathyroid Hormone/therapeutic use , Signal Transduction
13.
J Cell Physiol ; 205(1): 133-40, 2005 Oct.
Article in English | MEDLINE | ID: mdl-15828017

ABSTRACT

Apoptosis may play a role in osteoarthritis (OA). Apoptosis can proceed via two different pathways depending on the stimulus. However, both pathways converge upon the effector caspases, caspases-3 and -7. In some systems inhibition of caspases-3 and -7 can prevent apoptosis and may therefore have important therapeutic implications. To confirm this, apoptosis was induced in canine chondrocytes with mitomycin-c (MMC), either in the presence or absence of the general caspase inhibitor, Z-VAD FMK, or a specific caspase-3/7 inhibitor. Z-VAD FMK prevented MMC induced cell death. In contrast, inhibition of caspases-3 and -7 in the presence of MMC induced morphological changes that could be described as necrotic-like or paraptotic-like but did not prevent cell death. The addition of an inhibitor of caspase-8 or caspase-9 along with inhibitor of caspase-3/7 was required to reduce cell death. The morphological changes did not occur in the presence of the caspase-3/7 inhibitor alone and could be prevented by addition of Z-VAD FMK. These data lead to the conclusion that, if the apoptotic program cannot be completed, the cells are pushed into a necrotic or other nonapoptotic mode of death which may involve caspase-8 and/or caspase-9.


Subject(s)
Caspase Inhibitors , Chondrocytes/cytology , Chondrocytes/drug effects , Enzyme Inhibitors/pharmacology , Mitomycin/pharmacology , Animals , Apoptosis/drug effects , Caspase 3 , Caspase 7 , Caspases/metabolism , Cell Membrane Permeability/drug effects , Cell Shape/drug effects , Cells, Cultured , Chondrocytes/enzymology , Dogs , Microscopy, Electron, Transmission , Phenotype
14.
Curr Opin Pharmacol ; 4(3): 290-4, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15140422

ABSTRACT

The increase in marrow adipogenesis associated with osteoporosis and age-related osteopenia is well known clinically. However, we are only now beginning to understand the mechanisms that control the differentiation of mesenchymal stem cells to either osteoblasts or adipocytes. Recent work with gene silencing and overexpression has provided insight into critical pathways that determine the fate of these multipotential cells. One of these pathways - that of the nuclear hormone receptor peroxisome proliferator activated receptor-gamma - when activated, promotes adipogenesis and inhibits osteogenesis. This in vitro mechanism of action has been confirmed in vivo using ligands to this receptor. Discovery of this and other targets and pathways, such as Wnt signaling, notch/delta/jagged ligands and receptors, and RhoA gene expression, provides new insights into mesenchymal stem cell differentiation. These pathways provide exciting future pharmacological targets with which to enhance bone formation and therefore reduce the risk of fracture.


Subject(s)
Adipocytes/physiology , Aging/metabolism , Bone Marrow Cells/physiology , Osteoblasts/physiology , Osteoporosis/drug therapy , Receptors, Cytoplasmic and Nuclear/physiology , Transcription Factors/physiology , Adipocytes/metabolism , Animals , Female , Humans , Osteoblasts/metabolism , Osteoporosis/etiology , Osteoporosis/prevention & control , Receptors, Cytoplasmic and Nuclear/drug effects , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/drug effects , Transcription Factors/metabolism
15.
Endocrine ; 23(2-3): 183-8, 2004.
Article in English | MEDLINE | ID: mdl-15146099

ABSTRACT

Until recently, adipose tissue was considered to serve only as a triglyceride reservoir and was relegated to a passive endocrine role. With the discovery of leptin and other adipokines, adipose tissue is now recognized as an active participant in systemic metabolism. This review focuses on the complex relationship existing between adipose tissue and bone metabolism and differentiation. It explores the paradigms that have shaped the past decade's research and what these findings forecast for the future. Particular attention is given to the multipotent adult stem cell populations that reside within bone and fat. These adult stem cells have critical importance to the emerging field of tissue engineering and regenerative medicine.


Subject(s)
Adipose Tissue/anatomy & histology , Adipose Tissue/physiology , Bone and Bones/anatomy & histology , Bone and Bones/physiology , Adipose Tissue/cytology , Adipose Tissue/metabolism , Bone Marrow Cells/cytology , Bone and Bones/cytology , Bone and Bones/metabolism , Cell Differentiation , Humans , Mesoderm/cytology , Models, Biological , Osteoblasts/cytology , Stem Cells/cytology , Stromal Cells/cytology , Structure-Activity Relationship
16.
Expert Opin Ther Targets ; 6(6): 679-89, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12472380

ABSTRACT

Advances in genomics and proteomics have revolutionised the drug discovery process and target validation. Identification of novel therapeutic targets for chronic skeletal diseases is an extremely challenging process based on the difficulty of obtaining high-quality human diseased versus normal tissue samples. The quality of tissue and genomic information obtained from the sample is critical to identifying disease-related genes. Using a genomics-based approach, novel genes or genes with similar homology to existing genes can be identified from cDNA libraries generated from normal versus diseased tissue. High-quality cDNA libraries are prepared from uncontaminated homogeneous cell populations harvested from tissue sections of interest. Localised gene expression analysis and confirmation are obtained through in situ hybridisation or immunohistochemical studies. Cells overexpressing the recombinant protein are subsequently designed for primary cell-based high-throughput assays that are capable of screening large compound banks for potential hits. Afterwards, secondary functional assays are used to test promising compounds. The same overexpressing cells are used in the secondary assay to test protein activity and functionality as well as screen for small-molecule agonists or antagonists. Once a hit is generated, a structure-activity relationship of the compound is optimised for better oral bioavailability and pharmacokinetics allowing the compound to progress into development. Parallel efforts from proteomics, as well as genetics/transgenics, bioinformatics and combinatorial chemistry, and improvements in high-throughput automation technologies, allow the drug discovery process to meet the demands of the medicinal market. This review discusses and illustrates how different approaches are incorporated into the discovery and validation of novel targets and, consequently, the development of potentially therapeutic agents in the areas of osteoporosis and osteoarthritis. While current treatments exist in the form of hormone replacement therapy, antiresorptive and anabolic agents for osteoporosis, there are no disease-modifying therapies for the treatment of the most common human joint disease, osteoarthritis. A massive market potential for improved options with better safety and efficacy still remains. Therefore, the application of genomics and proteomics for both diseases should provide much needed novel therapeutic approaches to treating these major world health problems.


Subject(s)
Bone Diseases/drug therapy , Animals , Carrier Proteins , Caspases/physiology , Cathepsin K , Cathepsins/antagonists & inhibitors , Cysteine Proteinase Inhibitors/pharmacology , DNA-Binding Proteins/physiology , Drug Design , Drug Evaluation, Preclinical , Early Growth Response Protein 1 , Genomics , Glycoproteins/physiology , Humans , Immediate-Early Proteins/physiology , Membrane Glycoproteins , Mice , Mice, Knockout , Mice, Transgenic , Molecular Structure , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Osteoporosis/drug therapy , Osteoporosis/metabolism , Osteoprotegerin , Proteomics , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B , Receptors, Cytoplasmic and Nuclear/drug effects , Receptors, Cytoplasmic and Nuclear/physiology , Receptors, Tumor Necrosis Factor/physiology , Transcription Factors/physiology , Two-Hybrid System Techniques , src-Family Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...