Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
2.
Nat Commun ; 14(1): 4390, 2023 07 20.
Article in English | MEDLINE | ID: mdl-37474515

ABSTRACT

Replication Protein A (RPA) is a broadly conserved complex comprised of the RPA1, 2 and 3 subunits. RPA protects the exposed single-stranded DNA (ssDNA) during DNA replication and repair. Using structural modeling, we discover an inhibitor, JC-229, that targets RPA1 in Trypanosoma brucei, the causative parasite of African trypanosomiasis. The inhibitor is highly toxic to T. brucei cells, while mildly toxic to human cells. JC-229 treatment mimics the effects of TbRPA1 depletion, including DNA replication inhibition and DNA damage accumulation. In-vitro ssDNA-binding assays demonstrate that JC-229 inhibits the activity of TbRPA1, but not the human ortholog. Indeed, despite the high sequence identity with T. cruzi and Leishmania RPA1, JC-229 only impacts the ssDNA-binding activity of TbRPA1. Site-directed mutagenesis confirms that the DNA-Binding Domain A (DBD-A) in TbRPA1 contains a JC-229 binding pocket. Residue Serine 105 determines specific binding and inhibition of TbRPA1 but not T. cruzi and Leishmania RPA1. Our data suggest a path toward developing and testing highly specific inhibitors for the treatment of African trypanosomiasis.


Subject(s)
Chagas Disease , Trypanosoma brucei brucei , Trypanosomiasis, African , Animals , Humans , Trypanosoma brucei brucei/genetics , Replication Protein A/metabolism , DNA Replication , DNA, Single-Stranded/genetics , Protein Binding
3.
J Biol Chem ; 297(4): 101198, 2021 10.
Article in English | MEDLINE | ID: mdl-34534548

ABSTRACT

Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) fulfills various physiological roles that are unrelated to its glycolytic function. However, to date, the nonglycolytic function of GAPDH in trypanosomal parasites is absent from the literature. Exosomes secreted from Leishmania, like entire parasites, were found to have a significant impact on macrophage cell signaling and function, indicating cross talk with the host immune system. In this study, we demonstrate that the Leishmania GAPDH (LmGAPDH) protein is highly enriched within the extracellular vesicles (EVs) secreted during infection. To understand the function of LmGAPDH in EVs, we generated control, overexpressed, half-knockout (HKO), and complement cell lines. HKO cells displayed lower virulence compared with control cells when macrophages and BALB/c mice were infected with them, implying a crucial role for LmGAPDH in Leishmania infection and disease progression. Furthermore, upon infection of macrophages with HKO mutant Leishmania and its EVs, despite no differences in TNFA mRNA expression, there was a considerable increase in TNF-α protein expression compared with control, overexpressed, and complement parasites as determined by ELISA, RT-PCR, and immunoblot data. In vitro protein translation studies suggest that LmGAPDH-mediated TNF-α suppression occurs in a concentration-dependent manner. Moreover, mRNA binding assays also verified that LmGAPDH binds to the AU-rich 3'-UTR region of TNFA mRNA, limiting its production. Together, these findings confirmed that the LmGAPDH contained in EVs inhibits TNF-α expression in macrophages during infection via posttranscriptional repression.


Subject(s)
Extracellular Vesicles/enzymology , Gene Expression Regulation , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , Leishmania major/enzymology , Macrophages/metabolism , Protozoan Proteins/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Extracellular Vesicles/immunology , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/immunology , Leishmania major/immunology , Macrophages/immunology , Mice , Mice, Inbred BALB C , Protozoan Proteins/immunology , Tumor Necrosis Factor-alpha/immunology
4.
FEBS J ; 287(23): 5183-5195, 2020 12.
Article in English | MEDLINE | ID: mdl-32196942

ABSTRACT

Recently, we described the PAS domain-containing phosphoglycerate kinase (PGK) from Leishmania major (LmPAS-PGK) that shows acidic pH (5.5)-dependent optimum catalytic activity. The PAS domain of LmPAS-PGK is expected to regulate PGK activity during catalysis, but the mechanism of regulation by PAS domain at the molecular level is uncharacterized. In this work, we have utilized the full-length, PAS domain-deleted, and mutant enzymes to measure the enzymatic activity in the presence of divalent cation at various pH values. Catalytic activity measurement indicates that Mg2+ binding through PAS domain inhibits the PGK activity at pH 7.5, and this inhibition is withdrawn at pH 5.5. To identify the Mg2+ binding residues of the PAS domain, we exploited a systematic mutational analysis of all (four) His residues in the PAS domain for potential divalent cation binding. Replacement of His-57 with alanine resulted in depression in the presence of Mg2+ at pH 7.5, but H71A, H89A, and H111A showed similar characteristics with respect to the wild-type protein. Fluorescence and isothermal titration calorimetry studies revealed that H57 is responsible for Mg2+ binding in the absence of substrates. Thus, the protonated form of His57 at acidic pH 5.5 destabilizes the Mg2+ binding in the PAS domain, which is an essential requirement in the wild-type LmPAS-PGK for a conformational alteration in the sensor domain that, sequentially, activates the PGK domain, resulting in the synthesis of higher amounts of ATP.


Subject(s)
Leishmania major/enzymology , Magnesium/metabolism , Mutant Proteins/metabolism , Phosphoglycerate Kinase/metabolism , Protein Serine-Threonine Kinases/metabolism , Binding Sites , Catalysis , Hydrogen-Ion Concentration , Mutant Proteins/genetics , Mutation , Phosphoglycerate Kinase/genetics , Protein Serine-Threonine Kinases/genetics
5.
Biochem J ; 476(8): 1303-1321, 2019 04 30.
Article in English | MEDLINE | ID: mdl-30988012

ABSTRACT

Per-Arnt-Sim (PAS) domains are structurally conserved and present in numerous proteins throughout all branches of the phylogenetic tree. Although PAS domain-containing proteins are major players for the adaptation to environmental stimuli in both prokaryotic and eukaryotic organisms, these types of proteins are still uncharacterized in the trypanosomatid parasites, Trypanosome and Leishmania In addition, PAS-containing phosphoglycerate kinase (PGK) protein is uncharacterized in the literature. Here, we report a PAS domain-containing PGK (LmPAS-PGK) in the unicellular pathogen Leishmania The modeled structure of N-terminal of this protein exhibits four antiparallel ß sheets centrally flanked by α helices, which is similar to the characteristic signature of PAS domain. Activity measurements suggest that acidic pH can directly stimulate PGK activity. Localization studies demonstrate that the protein is highly enriched in the glycosome and its presence can also be seen in the lysosome. Gene knockout, overexpression and complement studies suggest that LmPAS-PGK plays a fundamental role in cell survival through autophagy. Furthermore, the knockout cells display a marked decrease in virulence when host macrophage and BALB/c mice were infected with them. Our work begins to clarify how acidic pH-dependent ATP generation by PGK is likely to function in cellular adaptability of Leishmania.


Subject(s)
Autophagosomes/immunology , Leishmania major , Macrophages , Models, Molecular , Phosphoglycerate Kinase , Protozoan Proteins , Animals , Leishmania major/genetics , Leishmania major/immunology , Leishmania major/pathogenicity , Macrophages/immunology , Macrophages/parasitology , Mice , Mice, Inbred BALB C , Phosphoglycerate Kinase/chemistry , Phosphoglycerate Kinase/deficiency , Phosphoglycerate Kinase/immunology , Protein Structure, Secondary , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Protozoan Proteins/immunology
6.
Biochem Biophys Res Commun ; 503(1): 371-377, 2018 09 03.
Article in English | MEDLINE | ID: mdl-29906460

ABSTRACT

Leishmania promastigotes have the ability to synthesize essential polyunsaturated fatty acids de novo and can grow in lipid free media. Recently, we have shown that NAD(P)H cytochrome b5 oxidoreductase (Ncb5or) enzyme in Leishmania acts as the redox partner for Δ12 fatty acid desaturase, which catalyses the conversion of oleate to linoleate. So far, the exact role of Leishmania derived linoleate synthesis is still incomplete in the literature. The viability assay by flow cytometry as well as microscopic studies suggests that linoleate is an absolute requirement for Leishmania promastigote survival in delipidated media. Western blot analysis suggested that infection with log phase linoleate deficient mutant (KO) results in increased level of NF-κBp65, IκB and IKKß phosphorylation in RAW264.7 cells. Similarly, the log phase KO infected RAW264.7 cells show dramatic increment of COX-2 expression and TNF-α secretion, compared to control or Ncb5or complement (CM) cell lines. The activation of inflammatory signaling pathways by KO mutant is significantly reduced when the RAW264.7 cells are pre-treated with BSA bound linoleate. Together, these findings confirmed that the leishmanial linoleate inhibits both COX-2 and TNF-α expression in macrophage via the inactivation of NF-κB signaling pathway. The stationary phase of KO promastigotes shows avirulence after infection in macrophages as well as inoculation into BALB/c mice; whereas CM cell lines show virulence. Collectively, these data provide strong evidence that de novo linoleate synthesis in Leishmania is an essential for parasite survival at extracellular promastigote stage as well as intracellular amastigote stage.


Subject(s)
Cytochrome-B(5) Reductase/genetics , Gene Deletion , Leishmania major/genetics , Leishmania major/pathogenicity , Leishmaniasis, Cutaneous/parasitology , Protozoan Proteins/genetics , Animals , Cyclooxygenase 2/genetics , Female , Gene Expression Regulation , Leishmania major/growth & development , Leishmaniasis, Cutaneous/genetics , Leishmaniasis, Cutaneous/pathology , Linoleic Acid/genetics , Mice , Mice, Inbred BALB C , RAW 264.7 Cells , Tumor Necrosis Factor-alpha/genetics , Virulence
7.
Am J Physiol Endocrinol Metab ; 311(2): E519-29, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27406741

ABSTRACT

Growth hormone (GH) plays an essential role in controlling somatic growth and in regulating multiple physiological processes in humans and other species. Insulin-like growth factor I (IGF-I), a conserved, secreted 70-amino acid peptide, is a critical mediator of many of the biological effects of GH. Previous studies have demonstrated that GH rapidly and potently promotes IGF-I gene expression in rodents and in some other mammals through the transcription factor STAT5b, leading to accumulation of IGF-I mRNAs and production of IGF-I. Despite this progress, very little is known about how GH or other trophic factors control human IGF1 gene expression, in large part because of the absence of any cellular model systems that robustly express IGF-I. Here, we have addressed mechanisms of regulation of human IGF-I by GH after generating cells in which the IGF1 chromosomal locus has been incorporated into a mouse cell line. Using this model, we found that physiological levels of GH rapidly stimulate human IGF1 gene transcription and identify several potential transcriptional enhancers in chromatin that bind STAT5b in a GH-regulated way. Each of the putative enhancers also activates a human IGF1 gene promoter in reconstitution experiments in the presence of the GH receptor, STAT5b, and GH. Thus we have developed a novel experimental platform that now may be used to determine how human IGF1 gene expression is controlled under different physiological and pathological conditions.


Subject(s)
Growth Hormone/metabolism , Insulin-Like Growth Factor I/genetics , RNA, Messenger/metabolism , Receptors, Somatotropin/metabolism , STAT5 Transcription Factor/metabolism , Animals , Cell Line , Gene Expression Regulation , Growth Hormone/pharmacology , Humans , Insulin-Like Growth Factor I/drug effects , Mice , Mice, Transgenic , Promoter Regions, Genetic , RNA, Messenger/drug effects , Receptors, Somatotropin/drug effects , Reverse Transcriptase Polymerase Chain Reaction , STAT5 Transcription Factor/drug effects , Transcription, Genetic
8.
Proc Natl Acad Sci U S A ; 113(9): 2472-7, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26884170

ABSTRACT

Ligands of the endothelial-enriched tunica interna endothelial cell kinase 2 (Tie2) are markedly imbalanced in severe infections associated with vascular leakage, yet regulation of the receptor itself has been understudied in this context. Here, we show that TIE2 gene expression may constitute a novel vascular barrier control mechanism in diverse infections. Tie2 expression declined rapidly in wide-ranging models of leak-associated infections, including anthrax, influenza, malaria, and sepsis. Forced Tie2 suppression sufficed to attenuate barrier function and sensitize endothelium to permeability mediators. Rapid reduction of pulmonary Tie2 in otherwise healthy animals attenuated downstream kinase signaling to the barrier effector vascular endothelial (VE)-cadherin and induced vascular leakage. Compared with wild-type littermates, mice possessing one allele of Tie2 suffered more severe vascular leakage and higher mortality in two different sepsis models. Common genetic variants that influence TIE2 expression were then sought in the HapMap3 cohort. Remarkably, each of the three strongest predicted cis-acting SNPs in HapMap3 was also associated with the risk of acute respiratory distress syndrome (ARDS) in an intensive care unit cohort of 1,614 subjects. The haplotype associated with the highest TIE2 expression conferred a 28% reduction in the risk of ARDS independent of other major clinical variables, including disease severity. In contrast, the most common haplotype was associated with both the lowest TIE2 expression and 31% higher ARDS risk. Together, the results implicate common genetic variation at the TIE2 locus as a determinant of vascular leak-related clinical outcomes from common infections, suggesting new tools to identify individuals at unusual risk for deleterious complications of infection.


Subject(s)
Capillary Permeability , Infections/physiopathology , Receptor, TIE-2/genetics , Animals , Endothelium, Vascular/physiopathology , Mice
9.
Physiol Genomics ; 47(11): 559-68, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26330488

ABSTRACT

Growth hormone (GH) plays a central role in regulating somatic growth and in controlling multiple physiological processes in humans and other vertebrates. A key agent in many GH actions is the secreted peptide, IGF-I. As established previously, GH stimulates IGF-I gene expression via the Stat5b transcription factor, leading to production of IGF-I mRNAs and proteins. However, the precise mechanisms by which GH-activated Stat5b promotes IGF-I gene transcription have not been defined. Unlike other GH-regulated genes, there are no Stat5b sites near either of the two IGF-I gene promoters. Although dispersed GH-activated Stat5b binding elements have been mapped in rodent Igf1 gene chromatin, it is unknown how these distal sites might function as potential transcriptional enhancers. Here we have addressed mechanisms of regulation of IGF-I gene transcription by GH by generating cell lines in which the rat Igf1 chromosomal locus has been incorporated into the mouse genome. Using these cells we find that physiological levels of GH rapidly and potently activate Igf1 gene transcription while stimulating physical interactions in chromatin between inducible Stat5b-binding elements and the Igf1 promoters. We have thus developed a robust experimental platform for elucidating how dispersed transcriptional enhancers control Igf1 gene expression under different biological conditions.


Subject(s)
Enhancer Elements, Genetic , Growth Hormone/physiology , Insulin-Like Growth Factor I/genetics , Animals , Male , Promoter Regions, Genetic , Rats , Rats, Sprague-Dawley , Transcription, Genetic/physiology , Transgenes
10.
Arch Biochem Biophys ; 579: 85-90, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26095616

ABSTRACT

The globin coupled heme containing adenylate cyclase from Leishmania major (HemAC-Lm) has two globin domains (globin-A and globin-B). Globin-B domain (210-360 amino acids) may guide the interaction between globin-A and adenylate cyclase domains for the regulation of catalysis. We investigated the role of globin-B domain in HemAC-Lm by constructing a series of mutants namely Δ209 (209 amino acids deleted), Δ360 (360 amino acids deleted), H161A, H311A and H311A-Δ209. Spectroscopic data suggest that the Δ209 and H311A-Δ209 proteins to be Fe(2+)-O2 form and apo form, respectively, indicating that His311 residue in the globin-B domain is crucial for heme binding in Δ209 protein. However, the H311A mutant is still of the Fe(2+)-O2 form whereas H161A mutant shows the apo form, indicating that only His161 residue in the globin-A domain is responsible for heme binding in full length enzyme. cAMP measurements suggest that the activities of Δ360 and Δ209 proteins were ∼10 and ∼1000 times lesser than full length enzyme, respectively, leading to the fact that globin-B domain inhibited catalysis rather than activation in absence of globin-A domain. These data suggest that the O2 bound globin-A domain in HemAC-Lm allows the best cooperation of the catalytic domain interactions to generate optimum cAMP.


Subject(s)
Adenylyl Cyclases/chemistry , Adenylyl Cyclases/metabolism , Escherichia coli/metabolism , Globins/chemistry , Globins/metabolism , Heme/chemistry , Heme/metabolism , Amino Acid Sequence , Binding Sites , Catalysis , Enzyme Activation , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , Structure-Activity Relationship , Substrate Specificity
11.
PLoS One ; 10(3): e0119577, 2015.
Article in English | MEDLINE | ID: mdl-25761062

ABSTRACT

BACKGROUND: Reactive oxygen species (ROS) are largely considered to be pathogenic to normal endothelial function in disease states such as sepsis. We hypothesized that Angiopoietin-1 (Angpt-1), an endogenous agonist of the endothelial-specific receptor, Tie-2, promotes barrier defense by activating NADPH oxidase (NOX) signaling. METHODS AND FINDINGS: Using primary human microvascular endothelial cells (HMVECs), we found that Angpt-1 stimulation induces phosphorylation of p47phox and a brief oxidative burst that is lost when chemical inhibitors of NOX activity or siRNA against the NOX component p47phox were applied. As a result, there was attenuated ROS activity, disrupted junctional contacts, enhanced actin stress fiber accumulation, and induced gap formation between confluent HMVECs. All of these changes were associated with weakened barrier function. The ability of Angpt-1 to prevent identical changes induced by inflammatory permeability mediators, thrombin and lipopolysaccharides (LPS), was abrogated by p47phox knockdown. P47phox was required for Angpt-1 to activate Rac1 and inhibit mediator-induced activation of the small GTPase RhoA. Finally, Angpt-1 gene transfer prevented vascular leakage in wildtype mice exposed to systemically administered LPS, but not in p47phox knock out (p47-/-) littermates. CONCLUSIONS: These results suggest an essential role for NOX signaling in Angpt-1-mediated endothelial barrier defense against mediators of systemic inflammation. More broadly, oxidants generated for signal transduction may have a barrier-promoting role in vascular endothelium.


Subject(s)
Angiopoietin-1/genetics , Angiopoietin-1/metabolism , Endothelium, Vascular/metabolism , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Oxidative Stress , Animals , Cells, Cultured , Endothelium, Vascular/cytology , Humans , Intercellular Junctions , Lipopolysaccharides/pharmacology , Membrane Glycoproteins/antagonists & inhibitors , Mice , NADPH Oxidase 2 , NADPH Oxidases/antagonists & inhibitors , Oxidative Stress/drug effects , Phosphorylation , Reactive Oxygen Species/metabolism , Thrombin/pharmacology
12.
PLoS One ; 9(3): e93040, 2014.
Article in English | MEDLINE | ID: mdl-24663486

ABSTRACT

Skeletal integrity is dependent on the coordinated actions of bone-forming osteoblasts and bone-resorbing osteoclasts, which recognize and respond to multiple environmental inputs. Here we have studied the roles in bone development and growth of Akt1 and Akt2, two closely related signaling proteins, by evaluating mice lacking either of these enzymes. Global deficiency of Akt1 but not Akt2 caused a reduction in whole body and femoral bone mineral density, in femoral cortical thickness and volume, and in trabecular thickness in both males and females when measured at 20-weeks of age, which was reflected in diminished femoral resistance to fracture. Haplo-deficiency of Akt1 in male mice also decreased femoral cortical and trabecular skeletal parameters, and reduced bone strength. Cell-based studies showed that genetic Akt1 deficiency diminished the rate of proliferation of osteoblast progenitors and impaired osteoclast differentiation in primary culture but that loss of Akt2 did not. Our results demonstrate differential effects of Akt1 and Akt2 on skeletal maturation and architecture through actions on both osteoblast and osteoclast precursors.


Subject(s)
Bone Density/physiology , Femur/enzymology , Osteoblasts/enzymology , Osteogenesis/physiology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Stem Cells/enzymology , Animals , Cell Differentiation/physiology , Female , Femur/cytology , Male , Mice , Mice, Mutant Strains , Osteoblasts/cytology , Proto-Oncogene Proteins c-akt/genetics , Stem Cells/cytology
13.
Proc Natl Acad Sci U S A ; 110(36): E3445-54, 2013 Sep 03.
Article in English | MEDLINE | ID: mdl-23946421

ABSTRACT

Stapled α-helical peptides have emerged as a promising new modality for a wide range of therapeutic targets. Here, we report a potent and selective dual inhibitor of MDM2 and MDMX, ATSP-7041, which effectively activates the p53 pathway in tumors in vitro and in vivo. Specifically, ATSP-7041 binds both MDM2 and MDMX with nanomolar affinities, shows submicromolar cellular activities in cancer cell lines in the presence of serum, and demonstrates highly specific, on-target mechanism of action. A high resolution (1.7-Å) X-ray crystal structure reveals its molecular interactions with the target protein MDMX, including multiple contacts with key amino acids as well as a role for the hydrocarbon staple itself in target engagement. Most importantly, ATSP-7041 demonstrates robust p53-dependent tumor growth suppression in MDM2/MDMX-overexpressing xenograft cancer models, with a high correlation to on-target pharmacodynamic activity, and possesses favorable pharmacokinetic and tissue distribution properties. Overall, ATSP-7041 demonstrates in vitro and in vivo proof-of-concept that stapled peptides can be developed as therapeutically relevant inhibitors of protein-protein interaction and may offer a viable modality for cancer therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Peptides/therapeutic use , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Area Under Curve , Binding, Competitive , Cell Line, Tumor , Crystallography, X-Ray , Female , HCT116 Cells , Humans , MCF-7 Cells , Male , Mice , Mice, Nude , Models, Molecular , Neoplasms/metabolism , Neoplasms/pathology , Peptides/chemistry , Peptides/metabolism , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacokinetics , Peptides, Cyclic/therapeutic use , Protein Binding , Protein Conformation , Protein Structure, Secondary , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Rats , Rats, Long-Evans , Xenograft Model Antitumor Assays
14.
Crit Care Med ; 40(11): 3034-41, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22890252

ABSTRACT

OBJECTIVE: : In sepsis, quiescent blood vessels become leaky and inflamed by mechanisms that are incompletely understood. We hypothesized that angiopoietin-2, a partial antagonist of the endothelium-stabilizing receptor Tie-2 secreted by endothelium, contributes to adverse outcomes in this disease. DESIGN: : Laboratory and animal research. SETTINGS: : Research laboratories and Emergency Department of Beth Israel Deaconess Medical Center, Boston, MA. SUBJECTS: : Angiopoietin-2 heterozygous mice, emergency department patients. MEASUREMENTS AND MAIN RESULTS: : Mice with one functional angiopoietin-2 allele developed milder kidney and lung injury, less tissue inflammation, and less vascular leakage compared to wild-type counterparts. Heterozygotes experienced >40% absolute survival advantage following two different models of sepsis (p = .004 and .018). In human subjects presenting to our emergency department with suspected infection (n = 270 combined), circulating angiopoietin-2 was markedly elevated within the first hour of clinical care. First-hour angiopoietin-2 concentrations were proportional to current disease severity (p < .0001), rose further over time in eventual nonsurvivors (p < .0001), and predicted the future occurrence of shock (p < .0001) or death (p < .0001) in the original cohort and an independent validation group. Finally, septic human serum disrupted the barrier function of microvascular endothelial cells, an effect fully neutralized by an angiopoietin-2 monoclonal antibody. CONCLUSIONS: : We conclude that angiopoietin-2 induction precedes and contributes to the adverse outcomes in sepsis, opening a new avenue for therapeutic investigation.


Subject(s)
Angiopoietin-2/physiology , Multiple Organ Failure/physiopathology , Sepsis/physiopathology , Adult , Aged , Aged, 80 and over , Angiopoietin-2/genetics , Angiopoietin-2/immunology , Animals , Female , Heterozygote , Humans , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Middle Aged , Multiple Organ Failure/mortality , Sepsis/mortality , Survival Analysis
15.
Proc Natl Acad Sci U S A ; 109(25): 10024-9, 2012 Jun 19.
Article in English | MEDLINE | ID: mdl-22665799

ABSTRACT

The anthrax lethal toxin (LT) enters host cells and enzymatically cleaves MAPKKs or MEKs. How these molecular events lead to death from anthrax remains poorly understood, but published reports suggest a direct effect of LT on vascular permeability. We have found that LT challenge in mice disrupts signaling through Tie-2, a tonically activated receptor tyrosine kinase in the endothelium. Genetic manipulations favoring Tie-2 activation enhanced interendothelial junctional contacts, prevented vascular leakage, and promoted survival following a lethal dose of LT. Cleavage of MEK1/2 was necessary for LT to induce endothelial barrier dysfunction, and activated Tie-2 signaled through the uncleaved fraction of MEKs to prevent LT's effects on the endothelium. Finally, primates infected with toxin-secreting Bacillus anthracis bacilli developed a rapid and marked imbalance in the endogenous ligands that signal Tie-2, similar to that seen in LT-challenged mice. Our results show that B. anthracis LT blunts signaling through Tie-2, thereby weakening the vascular barrier and contributing to lethality of the disease. Measurement of circulating Tie-2 ligands and manipulation of Tie-2 activity may represent future prognostic and therapeutic avenues for humans exposed to B. anthracis.


Subject(s)
Anthrax/physiopathology , Receptor, TIE-2/physiology , Angiopoietin-2/metabolism , Animals , Bacillus anthracis/metabolism , Cells, Cultured , Humans , Male , Mice , Mice, Inbred C57BL , Papio
16.
J Bone Miner Res ; 27(6): 1345-56, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22407846

ABSTRACT

Osteoporosis, the most common skeletal disorder, is characterized by low bone mineral density (BMD) and an increased risk of fragility fractures. BMD is the best clinical predictor of future osteoporotic fracture risk, but is a complex trait controlled by multiple environmental and genetic determinants with individually modest effects. Quantitative trait locus (QTL) mapping is a powerful method for identifying chromosomal regions encompassing genes involved in shaping complex phenotypes, such as BMD. Here we have applied QTL analysis to male and female genetically-heterogeneous F(2) mice derived from a cross between C57BL/6 and DBA/2 strains, and have identified 11 loci contributing to femoral BMD. Further analysis of a QTL on mouse chromosome 7 following the generation of reciprocal congenic strains has allowed us to determine that the high BMD trait, which tracks with the DBA/2 chromosome and exerts equivalent effects on male and female mice, is manifested by enhanced osteogenic differentiation of mesenchymal stem cells (MSCs) in vitro and by increased growth of metatarsal bones in short-term primary culture. An insertion/deletion DNA polymorphism in Ltbp4 exon 12 that causes the in-frame removal of 12 codons in the DBA/2-derived gene maps within 0.6 Mb of the marker most tightly linked to the QTL. LTBP4, one of four paralogous mouse proteins that modify the bioavailability of the transforming growth factor ß (TGF-ß) family of growth factors, is expressed in differentiating MSC-derived osteoblasts and in long bones, and reduced responsiveness to TGF-ß1 is observed in MSCs of mice homozygous for the DBA/2 chromosome 7. Taken together, our results identify a potential genetic and biochemical relationship between decreased TGF-ß1-mediated signaling and enhanced femoral BMD that may be regulated by a variant LTBP4 molecule.


Subject(s)
Bone and Bones/metabolism , Quantitative Trait Loci/genetics , Signal Transduction/genetics , Transforming Growth Factor beta1/metabolism , Animals , Bone Density/drug effects , Bone Density/genetics , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone Morphogenetic Protein 2/pharmacology , Bone and Bones/drug effects , Cell Differentiation/drug effects , Crosses, Genetic , Female , Femur/anatomy & histology , Femur/metabolism , Genetic Association Studies , Laboratories , Lod Score , Male , Metatarsal Bones/drug effects , Metatarsal Bones/growth & development , Mice , Mice, Congenic , Osteogenesis/drug effects , Osteogenesis/genetics , Phenotype , Quantitative Trait, Heritable , Rats , Signal Transduction/drug effects , Stromal Cells/cytology , Stromal Cells/drug effects , Stromal Cells/metabolism , Transforming Growth Factor beta1/genetics
17.
J Biol Chem ; 287(2): 1510-9, 2012 Jan 06.
Article in English | MEDLINE | ID: mdl-22117064

ABSTRACT

The six high-affinity insulin-like growth factor-binding proteins (IGFBPs) comprise a conserved family of secreted molecules that modulate IGF actions by regulating their half-life and access to signaling receptors, and also exert biological effects that are independent of IGF binding. IGFBPs are composed of cysteine-rich amino- (N-) and carboxyl- (C-) terminal domains, along with a cysteine-poor central linker segment. IGFBP-5 is the most conserved IGFBP, and contains 18 cysteines, but only 2 of 9 putative disulfide bonds have been mapped to date. Using a mass spectrometry (MS)-based strategy combining sequential electron transfer dissociation (ETD) and collision-induced dissociation (CID) steps, in which ETD fragmentation preferentially induces cleavage of disulfide bonds, and CID provides exact disulfide linkage assignments between liberated peptides, we now have definitively mapped 5 disulfide bonds in IGFBP-5. In addition, in conjunction with ab initio molecular modeling we are able to assign the other 4 disulfide linkages to within a GCGCCXXC motif that is conserved in five IGFBPs. Because of the nature of ETD fragmentation MS experiments were performed without chemical reduction of IGFBP-5. Our results not only establish a disulfide bond map of IGFBP-5 but also define a general approach that takes advantage of the specificity of ETD and the scalability of tandem MS, and the predictive power of ab initio molecular modeling to characterize unknown disulfide linkages in proteins.


Subject(s)
Disulfides/chemistry , Insulin-Like Growth Factor Binding Protein 5/chemistry , Models, Molecular , Peptide Mapping/methods , Amino Acid Motifs , Animals , Cell Line , Cysteine , Humans , Insulin-Like Growth Factor Binding Protein 5/genetics , Mass Spectrometry , Mice
18.
Mol Cell Biol ; 32(2): 490-500, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22064480

ABSTRACT

Maintaining optimal bone integrity, mass, and strength throughout adult life requires ongoing bone remodeling, which involves coordinated activity between actions of bone-resorbing osteoclasts and bone forming-osteoblasts. Osteoporosis is a disorder of remodeling in which bone resorption outstrips deposition, leading to diminished bone mass and an increased risk of fractures. Here we identify Akt1 as a unique signaling intermediate in osteoblasts that can control both osteoblast and osteoclast differentiation. Targeted knockdown of Akt1 in mouse primary bone marrow stromal cells or in a mesenchymal stem cell line or genetic knockout of Akt1 stimulated osteoblast differentiation secondary to increased expression of the osteogenic transcription factor Runx2. Despite enhanced osteoblast differentiation, coupled osteoclastogenesis in Akt1 deficiency was markedly inhibited, with reduced accumulation of specific osteoclast mRNAs and proteins and impaired fusion to form multinucleated osteoclasts, defects secondary to diminished production of receptor activator of NF-κB ligand (RANKL) and macrophage colony-stimulating factor (m-CSF), critical osteoblast-derived osteoclast differentiation factors. Delivery of recombinant lentiviruses encoding Akt1 but not Akt2 to Akt1-deficient osteoblast progenitors reversed the increased osteoblast differentiation and, by boosting accumulation of RANKL and m-CSF, restored normal osteoclastogenesis, as did the addition of recombinant RANKL to conditioned culture medium from Akt1-deficient osteoblasts. Our results support the idea that targeted inhibition of Akt1 could lead to therapeutically useful net bone acquisition, and they indicate that closely related Akt1 and Akt2 exert distinct effects on cellular differentiation pathways.


Subject(s)
Osteoblasts/cytology , Osteoclasts/cytology , Osteogenesis , Proto-Oncogene Proteins c-akt/metabolism , Animals , Bone Morphogenetic Protein 2/metabolism , Cell Differentiation , Cell Line , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/metabolism , Gene Knockdown Techniques , Mice , Mice, Inbred C57BL , Osteoblasts/metabolism , Osteoclasts/metabolism , Proto-Oncogene Proteins c-akt/genetics , RANK Ligand/metabolism , Signal Transduction
19.
Arterioscler Thromb Vasc Biol ; 31(11): 2643-52, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21885850

ABSTRACT

OBJECTIVE: IQ domain GTPase-activating protein 1 (IQGAP1) contributes to cytoskeletal network regulation in epithelial cells by its scaffolding properties and by binding the Rho GTPase Rac1 to maintain its activity. The functions of IQGAP1 in endothelial cells beyond angiogenesis remain unclear. We hypothesized that IQGAP1 participates in the regulation of endothelial barrier function. METHODS AND RESULTS: Silencing IQGAP1 in human microvascular endothelial cells resulted in a disruption of adherens junctions, formation of interendothelial gaps, and a reduction in barrier function. Furthermore, silencing of IQGAP1 abrogated the barrier enhancement effect of angiopoietin-1 (Angpt-1) and abolished the barrier-stabilizing effect of Angpt-1 on thrombin-stimulated cells. Coimmunoprecipitation detected binding of endogenous IQGAP1 with Rac1 at baseline that was stronger when Rac1 was activated and weaker when it was deactivated. Measurement of GTP-bound Rac1 revealed that Angpt-1 failed to activate Rac1 not only if IQGAP1 was silenced but also if cells were transfected with a mutant disabled in Rac1 binding (T1050AX2). Furthermore, a dominant-active Rac1 was sufficient to completely reverse the morphological and functional changes induced by reduction in IQGAP1. CONCLUSION: These experiments are the first demonstration of IQGAP1 regulating barrier function in any cell type. Further, our data show that Angpt-1 requires IQGAP1 as an indispensable activator of Rac1.


Subject(s)
Adherens Junctions/physiology , Angiopoietin-1/metabolism , Endothelium, Vascular/metabolism , Gap Junctions/physiology , rac1 GTP-Binding Protein/metabolism , ras GTPase-Activating Proteins/metabolism , Cell Membrane Permeability/physiology , Cells, Cultured , Endothelium, Vascular/cytology , Humans , RNA, Small Interfering/pharmacology , Signal Transduction/physiology , ras GTPase-Activating Proteins/antagonists & inhibitors , ras GTPase-Activating Proteins/drug effects
20.
Mol Cell Biol ; 30(4): 1018-27, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19995912

ABSTRACT

Mesenchymal stem cells are essential for repair of bone and other supporting tissues. Bone morphogenetic proteins (BMPs) promote commitment of these progenitors toward an osteoblast fate via functional interactions with osteogenic transcription factors, including Dlx3, Dlx5, and Runx2, and also can direct their differentiation into bone-forming cells. BMP-2-stimulated osteoblast differentiation additionally requires continual signaling from insulin-like growth factor (IGF)-activated pathways. Here we identify Akt2 as a critical mediator of IGF-regulated osteogenesis. Targeted knockdown of Akt2 in mouse primary bone marrow stromal cells or in a mesenchymal stem cell line, or genetic knockout of Akt2, did not interfere with BMP-2-mediated signaling but resulted in inhibition of osteoblast differentiation at an early step that preceded production of Runx2. In contrast, Akt1-deficient cells differentiated normally. Complete biochemical and morphological osteoblast differentiation was restored in cells lacking Akt2 by adenoviral delivery of Runx2 or by a recombinant lentivirus encoding wild-type Akt2. In contrast, lentiviral Akt1 was ineffective. Taken together, these observations define a specific role for Akt2 as a gatekeeper of osteogenic differentiation through regulation of Runx2 gene expression and indicate that the closely related Akt1 and Akt2 exert distinct effects on the differentiation of mesenchymal precursors.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Cell Differentiation , Osteoblasts/cytology , Osteoblasts/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Animals , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Gene Expression Regulation , Humans , Male , Mice , Osteogenesis , Proto-Oncogene Proteins c-akt/genetics
SELECTION OF CITATIONS
SEARCH DETAIL