Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Prostate ; 84(11): 1086-1088, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38678435

ABSTRACT

BACKGROUND: Prostate-specific membrane antigen (PSMA) is a biomarker and therapeutic target of high relevance in prostate cancer. Although upregulated PSMA expression is a well-documented feature of prostatic neoplasia in both humans and canids, to date humans are the only species known to express PSMA basally in the prostate. Thus, traditional laboratory animal species have limited utility for studying PSMA biology in the prostate or for predicting efficacy or toxicity of PSMA-targeted agents. METHODS: PSMA expression in human, macaque, and marmoset prostates was determined by immunohistochemistry, employing an antibody with validated cross-species reactivity in a PSMA-positive control tissue; kidney. RESULTS: We newly discover that the common marmoset endogenously expresses PSMA in non-diseased prostate, similar to humans, and thus may be a valuable preclinical model for researchers studying PSMA.


Subject(s)
Antigens, Surface , Callithrix , Glutamate Carboxypeptidase II , Prostate , Male , Animals , Glutamate Carboxypeptidase II/metabolism , Antigens, Surface/metabolism , Humans , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Immunohistochemistry
2.
Clin Exp Gastroenterol ; 16: 237-247, 2023.
Article in English | MEDLINE | ID: mdl-38090679

ABSTRACT

Background: Prostate-specific membrane antigen (PSMA) is highly and specifically upregulated in active-inflamed mucosa of patients with inflammatory bowel disease (IBD). We hypothesized that this upregulation would be detectable using a PSMA-targeted positron emission tomography/computed tomography (PET/CT) imaging agent, [18F]DCFPyL, enabling non-invasive visualization of inflammation. A noninvasive means of detecting active inflammation would have high clinical value in localization and management of IBD. Study: We performed [18F]DCFPyL imaging in three IBD patients with active disease. Abnormally increased gastrointestinal [18F]DCFPyL uptake was observed in areas with endoscopic, histologic, and immunohistochemical inflammation, demonstrating partial overlap of segments of bowel with abnormal [18F]DCFPyL uptake and active inflammation. Conclusion: This study demonstrates that PSMA-targeted [18F]DCFPyL PET can effectively detect regions of inflamed mucosa in patients with IBD, suggesting its utility as a non-invasive imaging agent to assess location, extent, and disease activity in IBD.

3.
Mol Cancer Ther ; 22(12): 1390-1403, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37616542

ABSTRACT

Malignant peripheral nerve sheath tumors (MPNST) are highly aggressive soft-tissue sarcomas that arise from neural tissues and carry a poor prognosis. Previously, we found that the glutamine amidotransferase inhibitor JHU395 partially impeded tumor growth in preclinical models of MPNST. JHU395 inhibits de novo purine synthesis in human MPNST cells and murine tumors with partial decreases in purine monophosphates. On the basis of prior studies showing enhanced efficacy when glutamine amidotransferase inhibition was combined with the antimetabolite 6-mercaptopurine (6-MP), we hypothesized that such a combination would be efficacious in MPNST. Given the known toxicity associated with 6-MP, we set out to develop a more efficient and well-tolerated drug that targets the purine salvage pathway. Here, we report the discovery of Pro-905, a phosphoramidate protide that delivered the active nucleotide antimetabolite thioguanosine monophosphate (TGMP) to tumors over 2.5 times better than equimolar 6-MP. Pro-905 effectively prevented the incorporation of purine salvage substrates into nucleic acids and inhibited colony formation of human MPNST cells in a dose-dependent manner. In addition, Pro-905 inhibited MPNST growth and was well-tolerated in both human patient-derived xenograft (PDX) and murine flank MPNST models. When combined with JHU395, Pro-905 enhanced the colony formation inhibitory potency of JHU395 in human MPNST cells and augmented the antitumor efficacy of JHU395 in mice. In summary, the dual inhibition of the de novo and purine salvage pathways in preclinical models may safely be used to enhance therapeutic efficacy against MPNST.


Subject(s)
Nerve Sheath Neoplasms , Neurofibrosarcoma , Humans , Animals , Mice , Glutamine , Cell Line, Tumor , Antimetabolites/therapeutic use , Nerve Sheath Neoplasms/drug therapy
4.
Sci Transl Med ; 15(708): eabn7491, 2023 08 09.
Article in English | MEDLINE | ID: mdl-37556558

ABSTRACT

There is an urgent need to develop therapeutics for inflammatory bowel disease (IBD) because up to 40% of patients with moderate-to-severe IBD are not adequately controlled with existing drugs. Glutamate carboxypeptidase II (GCPII) has emerged as a promising therapeutic target. This enzyme is minimally expressed in normal ileum and colon, but it is markedly up-regulated in biopsies from patients with IBD and preclinical colitis models. Here, we generated a class of GCPII inhibitors designed to be gut-restricted for oral administration, and we interrogated efficacy and mechanism using in vitro and in vivo models. The lead inhibitor, (S)-IBD3540, was potent (half maximal inhibitory concentration = 4 nanomolar), selective, gut-restricted (AUCcolon/plasma > 50 in mice with colitis), and efficacious in acute and chronic rodent colitis models. In dextran sulfate sodium-induced colitis, oral (S)-IBD3540 inhibited >75% of colon GCPII activity, dose-dependently improved gross and histologic disease, and markedly attenuated monocytic inflammation. In spontaneous colitis in interleukin-10 (IL-10) knockout mice, once-daily oral (S)-IBD3540 initiated after disease onset improved disease, normalized colon histology, and attenuated inflammation as evidenced by reduced fecal lipocalin 2 and colon pro-inflammatory cytokines/chemokines, including tumor necrosis factor-α and IL-17. Using primary human colon epithelial air-liquid interface monolayers to interrogate the mechanism, we further found that (S)-IBD3540 protected against submersion-induced oxidative stress injury by decreasing barrier permeability, normalizing tight junction protein expression, and reducing procaspase-3 activation. Together, this work demonstrated that local inhibition of dysregulated gastrointestinal GCPII using the gut-restricted, orally active, small-molecule (S)-IBD3540 is a promising approach for IBD treatment.


Subject(s)
Colitis , Glutamate Carboxypeptidase II , Inflammatory Bowel Diseases , Animals , Humans , Mice , Colitis/drug therapy , Colitis/metabolism , Colon/pathology , Cytokines/metabolism , Disease Models, Animal , Glutamate Carboxypeptidase II/antagonists & inhibitors , Inflammation/pathology , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/pathology , Mice, Inbred C57BL
5.
J Safety Res ; 84: 18-23, 2023 02.
Article in English | MEDLINE | ID: mdl-36868645

ABSTRACT

INTRODUCTION: Older vehicles, commonly referred to as "classic," "vintage," or "historic" vehicles (CVH), share the roadways with newer vehicles. Older vehicles lacking safety systems likely come with an increased risk of fatality, however there is no study examining the typical conditions for crashes involving CVH. METHOD: This study utilized information from crashes occurring in 2012 to 2019 to estimate fatal crash rates for vehicles grouped by model year deciles. Data from crashes documented in the National Highway Traffic Safety Administration's (NHTSA) FARS and GES/CRSS data sets were utilized to examine roadway, temporal, and crash types for passenger vehicles produced in 1970 or earlier (CVH). RESULTS: These data show CVH crashes are rare (<1% of crashes), but carry a relative risk of fatality from 6.70 (95th CI: 5.44-8.26) for impacts with other vehicles, which was the most common crash, to 9.53 (7.28-12.47) for rollovers. Most crashes occurred in dry weather, typically during summer, in rural areas, most frequently on two lane roads, and in areas with speed limits between 30 and 55 mph. Factors associated with fatality for occupants in CVH included alcohol use, lack of seat belt use, and older age. CONCLUSIONS AND PRACTICAL APPLICATIONS: Crashes involving a CVH are a rare event but have catastrophic consequences when they do occur. Regulations that limit driving to daylight hours may lower the risk of crash involvement, and safety messaging to promote belt use and sober driving may also help. Additionally, as new "smart" vehicles are developed, engineers should keep in mind that older vehicles remain on the roadway. New driving technologies will need to safely interact with these older, less safe vehicles.


Subject(s)
Alcohol Drinking , Automobile Driving , Humans , Engineering , Seasons , Seat Belts
6.
Sci Adv ; 8(46): eabq5925, 2022 11 18.
Article in English | MEDLINE | ID: mdl-36383674

ABSTRACT

6-Diazo-5-oxo-l-norleucine (DON) is a glutamine antagonist that suppresses cancer cell metabolism but concurrently enhances the metabolic fitness of tumor CD8+ T cells. DON showed promising efficacy in clinical trials; however, its development was halted by dose-limiting gastrointestinal (GI) toxicities. Given its clinical potential, we designed DON peptide prodrugs and found DRP-104 [isopropyl(S)-2-((S)-2-acetamido-3-(1H-indol-3-yl)-propanamido)-6-diazo-5-oxo-hexanoate] that was preferentially bioactivated to DON in tumor while bioinactivated to an inert metabolite in GI tissues. In drug distribution studies, DRP-104 delivered a prodigious 11-fold greater exposure of DON to tumor versus GI tissues. DRP-104 affected multiple metabolic pathways in tumor, including decreased glutamine flux into the TCA cycle. In efficacy studies, both DRP-104 and DON caused complete tumor regression; however, DRP-104 had a markedly improved tolerability profile. DRP-104's effect was CD8+ T cell dependent and resulted in robust immunologic memory. DRP-104 represents a first-in-class prodrug with differential metabolism in target versus toxicity tissue. DRP-104 is now in clinical trials under the FDA Fast Track designation.


Subject(s)
Neoplasms , Prodrugs , Humans , Prodrugs/pharmacology , Prodrugs/therapeutic use , Diazooxonorleucine/pharmacology , Diazooxonorleucine/therapeutic use , Glutamine/metabolism , CD8-Positive T-Lymphocytes/metabolism , Neoplasms/drug therapy , Enzyme Inhibitors/therapeutic use
9.
Neurochem Res ; 45(6): 1256-1267, 2020 Jun.
Article in English | MEDLINE | ID: mdl-31749072

ABSTRACT

In tribute to our friend and colleague Michael Robinson, we review his involvement in the identification, characterization and localization of the metallopeptidase glutamate carboxypeptidase II (GCPII), originally called NAALADase. While Mike was characterizing NAALADase in the brain, the protein was independently identified by other laboratories in human prostate where it was termed prostate specific membrane antigen (PSMA) and in the intestines where it was named Folate Hydrolase 1 (FOLH1). It was almost a decade to establish that NAALADase, PSMA, and FOLH1 are encoded by the same gene. The enzyme has emerged as a therapeutic target outside of the brain, with the most notable progress made in the treatment of prostate cancer and inflammatory bowel disease (IBD). PSMA-PET imaging with high affinity ligands is proving useful for the clinical diagnosis and staging of prostate cancer. A molecular radiotherapy based on similar ligands is in trials for metastatic castration-resistant prostate cancer. New PSMA inhibitor prodrugs that preferentially block kidney and salivary gland versus prostate tumor enzyme may improve the clinical safety of this radiotherapy. The wide clinical use of PSMA-PET imaging in prostate cancer has coincidentally led to clinical documentation of GCPII upregulation in a wide variety of tumors and inflammatory diseases, likely associated with angiogenesis. In IBD, expression of the FOLH1 gene that codes for GCPII is strongly upregulated, as is the enzymatic activity in diseased patient biopsies. In animal models of IBD, GCPII inhibitors show substantial efficacy, suggesting potential theranostic use of GCPII ligands for IBD.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain/drug effects , Brain/metabolism , Glutamate Carboxypeptidase II/antagonists & inhibitors , Glutamate Carboxypeptidase II/metabolism , Prodrugs/therapeutic use , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/metabolism , Humans , Male , Prodrugs/chemistry , Prodrugs/pharmacology , Prostate/drug effects , Prostate/metabolism
10.
Obesity (Silver Spring) ; 25(4): 689-696, 2017 04.
Article in English | MEDLINE | ID: mdl-28236433

ABSTRACT

OBJECTIVE: Obesity exists with and without accompanying cardiometabolic disease, termed metabolically unhealthy obesity (MUO) and healthy obesity (MHO), respectively. Underlying differences in the ability of subcutaneous (SQ) fat to respond to nutrient excess are emerging as a key pathway. This study aimed to document the first spontaneous animal model of MHO and MUO and differences in SQ adipose tissue. METHODS: Vervet monkeys (Chlorocebus aethiops; N = 171) were screened for metabolic syndrome. A subset of MHO and MUO monkeys (n = 6/group) had SQ fat biopsies collected for histological evaluations and examination of key mitochondrial proteins. RESULTS: Obesity was seen in 20% of monkeys, and within this population, 31% were healthy, which mirrors human prevalence estimates. MUO monkeys had more than 60% lower adiponectin concentrations despite similar fat cell size, uncoupling protein 3, and activated macrophage abundance. However, alternatively activated/anti-inflammatory macrophages were 70% lower. Deficiencies of 50% or more in mitochondrial quality control regulators and selected mitochondrial fission and fusion markers were observed in the SQ fat of MUO monkeys despite comparable mitochondrial content. CONCLUSIONS: A novel and translatable spontaneously obese animal model of MHO and MUO, occurring independently of dietary factors, was characterized. Differences in mitochondrial quality and inflammatory cell populations of subcutaneous fat may underpin divergent metabolic health.


Subject(s)
Metabolic Syndrome/physiopathology , Mitochondria/metabolism , Obesity/physiopathology , Subcutaneous Fat/metabolism , Adiponectin/analysis , Animals , Biomarkers/analysis , Chlorocebus aethiops , Disease Models, Animal , Macrophage Activation , Macrophages/metabolism , Subcutaneous Fat/cytology , Uncoupling Protein 3/analysis
11.
Development ; 143(15): 2818-28, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27385010

ABSTRACT

The membrane-anchored serine proteases prostasin (PRSS8) and matriptase (ST14) initiate a cell surface proteolytic pathway essential for epithelial function. Mice expressing only catalytically inactive prostasin are viable, unlike prostasin null mice, indicating that at least some prostasin functions are non-proteolytic. Here we used knock-in mice expressing catalytically inactive prostasin (Prss8(Ki/Ki)) to show that the physiological and pathological functions of prostasin vary in their dependence on its catalytic activity. Whereas prostasin null mice exhibited partial embryonic and complete perinatal lethality, Prss8(Ki/Ki) mice displayed normal prenatal and postnatal survival. Unexpectedly, catalytically inactive prostasin caused embryonic lethality in mice lacking its cognate inhibitors HAI-1 (SPINT1) or HAI-2 (SPINT2). Proteolytically inactive prostasin, unlike the wild-type protease, was unable to activate matriptase during placentation. Surprisingly, all essential functions of prostasin in embryonic and postnatal development were compensated for by loss of HAI-1, indicating that prostasin is only required for mouse development and overall viability in the presence of this inhibitor. This study expands our knowledge of non-proteolytic functions of membrane-anchored serine proteases and provides unexpected new data on the mechanistic interactions between matriptase and prostasin in the context of epithelial development.


Subject(s)
Serine Endopeptidases/metabolism , Serine Proteases/metabolism , Animals , Female , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Placentation , Pregnancy , Proteinase Inhibitory Proteins, Secretory , Proteolysis , Serine Endopeptidases/genetics , Serine Proteases/genetics
13.
Sci Rep ; 5: 16267, 2015 Nov 20.
Article in English | MEDLINE | ID: mdl-26584669

ABSTRACT

Anthrax lethal toxin (LT) is an A-B type toxin secreted by Bacillus anthracis, consisting of the cellular binding moiety, protective antigen (PA), and the catalytic moiety, lethal factor (LF). To target cells, PA binds to cell-surface receptors and is then proteolytically processed forming a LF-binding competent PA oligomer where each LF binding site is comprised of three subsites on two adjacent PA monomers. We previously generated PA-U2-R200A, a urokinase-activated PA variant with LF-binding subsite II residue Arg200 mutated to Ala, and PA-L1-I210A, a matrix metalloproteinase-activated PA variant with subsite III residue Ile210 mutated to Ala. PA-U2-R200A and PA-L1-I210A displayed reduced cytotoxicity when used singly. However, when combined, they formed LF-binding competent heterogeneous oligomers by intermolecular complementation, and achieved high specificity in tumor targeting. Nevertheless, each of these proteins, in particular PA-L1-I210A, retained residual LF-binding ability. In this work, we screened a library containing all possible amino acid substitutions for LF-binding site to find variants with activity strictly dependent upon intermolecular complementation. PA-I207R was identified as an excellent replacement for the original clockwise-side variant, PA-I210A. Consequently, the new combination of PA-L1-I207R and PA-U2-R200A showed potent anti-tumor activity and low toxicity, exceeding the performance of the original combination, and warranting further investigation.


Subject(s)
Antigens, Bacterial/pharmacology , Bacterial Toxins/pharmacology , Macrophages/drug effects , Mutant Proteins/pharmacology , Neoplasms, Experimental/drug therapy , Animals , Antigens, Bacterial/genetics , Bacterial Toxins/genetics , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Female , Macrophages/cytology , Male , Mice, Inbred C57BL , Mutant Proteins/genetics , Mutation, Missense , Neoplasms, Experimental/pathology , Time Factors , Tumor Burden/drug effects
14.
PLoS Genet ; 10(7): e1004470, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25078604

ABSTRACT

The development of eutherian mammalian embryos is critically dependent on the selective bi-directional transport of molecules across the placenta. Here, we uncover two independent and partially redundant protease signaling pathways that include the membrane-anchored serine proteases, matriptase and prostasin, and the G protein-coupled receptor PAR-2 that mediate the establishment of a functional feto-maternal barrier. Mice with a combined matriptase and PAR-2 deficiency do not survive to term and the survival of matriptase-deficient mice heterozygous for PAR-2 is severely diminished. Embryos with the combined loss of PAR-2 and matriptase or PAR-2 and the matriptase partner protease, prostasin, uniformly die on or before embryonic day 14.5. Despite the extensive co-localization of matriptase, prostasin, and PAR-2 in embryonic epithelia, the overall macroscopic and histological analysis of the double-deficient embryos did not reveal any obvious developmental abnormalities. In agreement with this, the conditional deletion of matriptase from the embryo proper did not affect the prenatal development or survival of PAR-2-deficient mice, indicating that the critical redundant functions of matriptase/prostasin and PAR-2 are limited to extraembryonic tissues. Indeed, placentas of the double-deficient animals showed decreased vascularization, and the ability of placental epithelium to establish a functional feto-maternal barrier was severely diminished. Interestingly, molecular analysis suggested that the barrier defect was associated with a selective deficiency in the expression of the tight junction protein, claudin-1. Our results reveal unexpected complementary roles of matriptase-prostasin- and PAR-2-dependent proteolytic signaling in the establishment of placental epithelial barrier function and overall embryonic survival.


Subject(s)
Mother-Child Relations , Oligopeptides/genetics , Placentation , Serine Endopeptidases/genetics , Animals , Cell Survival/genetics , Claudin-1/metabolism , Female , Mice , Morphogenesis/genetics , Oligopeptides/metabolism , Placenta/metabolism , Pregnancy , Serine Endopeptidases/metabolism , Signal Transduction/genetics
15.
Toxicol Appl Pharmacol ; 279(2): 220-9, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24971906

ABSTRACT

We have previously designed and characterized versions of anthrax lethal toxin that are selectively cytotoxic in the tumor microenvironment and which display broad and potent anti-tumor activities in vivo. Here, we have performed the first direct comparison of the safety and efficacy of three engineered anthrax lethal toxin variants requiring activation by either matrix-metalloproteinases (MMPs), urokinase plasminogen activator (uPA) or co-localized MMP/uPA activities. C57BL/6J mice were challenged with six doses of engineered toxins via intraperitoneal (I.P.) or intravenous (I.V.) dose routes to determine the maximum tolerated dose for six administrations (MTD6) and dose-limiting toxicities. Efficacy was evaluated using the B16-BL6 syngraft model of melanoma; mice bearing established tumors were treated with six I.P. doses of toxin and tumor measurements and immunohistochemistry, paired with terminal blood work, were used to elaborate upon the anti-tumor mechanism and relative efficacy of each variant. We found that MMP-, uPA- and dual MMP/uPA-activated anthrax lethal toxins exhibited the same dose-limiting toxicity; dose-dependent GI toxicity. In terms of efficacy, all three toxins significantly reduced primary B16-BL6 tumor burden, ranging from 32% to 87% reduction, and they also delayed disease progression as evidenced by dose-dependent normalization of blood work values. While target organ toxicity and effective doses were similar amongst the variants, the dual MMP/uPA-activated anthrax lethal toxin exhibited the highest I.P. MTD6 and was 1.5-3-fold better tolerated than the single MMP- and uPA-activated toxins. Overall, we demonstrate that this dual MMP/uPA-activated anthrax lethal toxin can be administered safely and is highly effective in a preclinical model of melanoma. This modified bacterial cytotoxin is thus a promising candidate for further clinical development and evaluation for use in treating human cancers.


Subject(s)
Antigens, Bacterial/pharmacology , Antineoplastic Agents/pharmacology , Bacterial Toxins/pharmacology , Melanoma, Experimental/drug therapy , Prodrugs/pharmacology , Protein Engineering , Skin Neoplasms/drug therapy , Animals , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/genetics , Antigens, Bacterial/metabolism , Antigens, Bacterial/toxicity , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/metabolism , Antineoplastic Agents/toxicity , Bacterial Toxins/administration & dosage , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Bacterial Toxins/toxicity , Biomarkers, Tumor/blood , Dose-Response Relationship, Drug , Female , Injections, Intraperitoneal , Injections, Intravenous , Matrix Metalloproteinases/metabolism , Maximum Tolerated Dose , Melanoma, Experimental/blood , Melanoma, Experimental/enzymology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Prodrugs/administration & dosage , Prodrugs/metabolism , Prodrugs/toxicity , Skin Neoplasms/blood , Skin Neoplasms/enzymology , Skin Neoplasms/pathology , Time Factors , Tumor Burden/drug effects , Urokinase-Type Plasminogen Activator/metabolism
16.
J Biol Chem ; 289(21): 14740-9, 2014 May 23.
Article in English | MEDLINE | ID: mdl-24706745

ABSTRACT

The membrane-anchored serine protease prostasin (CAP1/PRSS8) is part of a cell surface proteolytic cascade that is essential for epithelial barrier formation and homeostasis. Here, we report the surprising finding that prostasin executes these functions independent of its own enzymatic activity. Prostasin null (Prss8(-/-)) mice lack barrier formation and display fatal postnatal dehydration. In sharp contrast, mice homozygous for a point mutation in the Prss8 gene, which causes the substitution of the active site serine within the catalytic histidine-aspartate-serine triad with alanine and renders prostasin catalytically inactive (Prss8(Cat-/Cat-) mice), develop barrier function and are healthy when followed for up to 20 weeks. This striking difference could not be explained by genetic modifiers or by maternal effects, as these divergent phenotypes were displayed by Prss8(-/-) and Prss8(Cat-/Cat-) mice born within the same litter. Furthermore, Prss8(Cat-/Cat-) mice were able to regenerate epidermal covering following cutaneous wounding. This study provides the first demonstration that essential in vivo functions of prostasin are executed by a non-enzymatic activity of this unique membrane-anchored serine protease.


Subject(s)
Cell Membrane/enzymology , Epidermis/enzymology , Homeostasis/physiology , Serine Endopeptidases/metabolism , Animals , Animals, Newborn , Biocatalysis , Blotting, Western , Body Weight/genetics , Epidermis/growth & development , Epidermis/metabolism , Homeostasis/genetics , Homozygote , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Point Mutation , Serine Endopeptidases/genetics
17.
J Med Chem ; 56(21): 8339-51, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-24044354

ABSTRACT

Bioactive peptides have evolved to optimally fulfill specific biological functions, a fact which has long attracted attention for their use as therapeutic agents. While there have been some recent commercial successes fostered in part by advances in large-scale peptide synthesis, development of peptides as therapeutic agents has been significantly impeded by their inherent susceptibility to protease degradation in the bloodstream. Here we report that incorporation of specially designed amino acid analogues at the P1' position, directly C-terminal of the enzyme cleavage site, renders peptides, including glucagon-like peptide-1 (7-36) amide (GLP-1) and six other examples, highly resistant to serine protease degradation without significant alteration of their biological activity. We demonstrate the applicability of the method to a variety of proteases, including dipeptidyl peptidase IV (DPP IV), dipeptidyl peptidase 8 (DPP8), fibroblast activation protein α (FAPα), α-lytic protease (αLP), trypsin, and chymotrypsin. In summary, the "P1' modification" represents a simple, general, and highly adaptable method of generating enzymatically stable peptide-based therapeutics.


Subject(s)
Dipeptidyl Peptidase 4/metabolism , Peptides/pharmacology , Serine Proteinase Inhibitors/pharmacology , Animals , Dose-Response Relationship, Drug , Female , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Molecular Structure , Peptides/chemical synthesis , Peptides/chemistry , Serine Proteinase Inhibitors/chemical synthesis , Serine Proteinase Inhibitors/chemistry , Structure-Activity Relationship , Substrate Specificity
18.
J Cell Biol ; 202(6): 951-66, 2013 Sep 16.
Article in English | MEDLINE | ID: mdl-24019537

ABSTRACT

Tissue remodeling processes critically depend on the timely removal and remodeling of preexisting collagen scaffolds. Nevertheless, many aspects related to the turnover of this abundant extracellular matrix component in vivo are still incompletely understood. We therefore took advantage of recent advances in optical imaging to develop an assay to visualize collagen turnover in situ and identify cell types and molecules involved in this process. Collagen introduced into the dermis of mice underwent cellular endocytosis in a partially matrix metalloproteinase-dependent manner and was subsequently routed to lysosomes for complete degradation. Collagen uptake was predominantly executed by a quantitatively minor population of M2-like macrophages, whereas more abundant Col1a1-expressing fibroblasts and Cx3cr1-expressing macrophages internalized collagen at lower levels. Genetic ablation of the collagen receptors mannose receptor (Mrc1) and urokinase plasminogen activator receptor-associated protein (Endo180 and Mrc2) impaired this intracellular collagen degradation pathway. This study demonstrates the importance of receptor-mediated cellular uptake to collagen turnover in vivo and identifies a key role of M2-like macrophages in this process.


Subject(s)
Collagen Type I/physiology , Collagen/metabolism , Macrophages/metabolism , Membrane Glycoproteins/physiology , Receptors, Cell Surface/physiology , Receptors, Chemokine/physiology , Animals , Apoptosis , Blotting, Western , CX3C Chemokine Receptor 1 , Cell Proliferation , Cells, Cultured , Collagen Type I, alpha 1 Chain , Endocytosis/physiology , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Immunoenzyme Techniques , Lysosomes/metabolism , Macrophages/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Immunologic , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
19.
J Biol Chem ; 288(26): 19028-39, 2013 Jun 28.
Article in English | MEDLINE | ID: mdl-23673661

ABSTRACT

Matriptase and prostasin are part of a cell surface proteolytic pathway critical for epithelial development and homeostasis. Here we have used a reconstituted cell-based system and transgenic mice to investigate the mechanistic interrelationship between the two proteases. We show that matriptase and prostasin form a reciprocal zymogen activation complex with unique features. Prostasin serves as a critical co-factor for matriptase activation. Unexpectedly, however, prostasin-induced matriptase activation requires neither prostasin zymogen conversion nor prostasin catalytic activity. Prostasin zymogen conversion to active prostasin is dependent on matriptase but does not require matriptase zymogen conversion. Consistent with these findings, wild type prostasin, activation cleavage site-mutated prostasin, and catalytically inactive prostasin all were biologically active in vivo when overexpressed in the epidermis of transgenic mice, giving rise to a severe skin phenotype. Our finding of non-enzymatic stimulation of matriptase activation by prostasin and activation of prostasin by the matriptase zymogen provides a tentative mechanistic explanation for several hitherto unaccounted for genetic and biochemical observations regarding these two membrane-anchored serine proteases and their downstream targets.


Subject(s)
Enzyme Activation , Enzyme Precursors/chemistry , Serine Endopeptidases/chemistry , Allosteric Site , Animals , Binding Sites , Caco-2 Cells , Catalysis , Epithelial Cells/enzymology , Female , HEK293 Cells , Humans , Male , Mice , Mice, Transgenic , Peptide Hydrolases/chemistry , Phenotype
20.
Biochem Biophys Res Commun ; 422(3): 393-7, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22575514

ABSTRACT

Capillary morphogenesis protein-2 (CMG2) functions as an anthrax toxin receptor that plays an essential role in anthrax pathogenesis. Although mutations in CMG2 have been identified to cause two human autosomal recessive disorders, Juvenile Hyaline Fibromatosis and Infantile Systemic Hyalinosis, both characterized by excess hyaline material deposition in connective tissues, the physiologic function of CMG2 remains elusive. To study the roles of CMG2 in normal physiology, here we performed detailed histological analyses of the CMG2-null mice we generated previously. While no morphological or histological defects were observed in CMG2(-/-) male mice, CMG2(-/-) female mice were unable to produce any offspring due to a defect in parturition. We found that deletion of CMG2 resulted in a diffuse deposition of collagen within the myometrium of CMG2(-/-) females, causing remarkable morphological changes to their uteri. This collagen accumulation also led to loss of smooth muscle cells in the myometrium of CMG2(-/-) mice, apparently disabling uterine contractile function during parturition. As a consequence, even though pregnant CMG2(-/-) mice were able to carry the gestation to full term, they were unable to deliver pups. However, the fully-developed fetuses could be successfully delivered by Cesarean section and survived to adulthood when fostered. Our results demonstrate that CMG2 is not required for normal mouse embryonic development but is indispensable for murine parturition. In parallel to its role in anthrax toxin binding and internalization, herein we provide evidence that CMG2 may function as a collagen receptor which is essential for maintaining collagen homeostasis in the uterus.


Subject(s)
Collagen/physiology , Homeostasis , Parturition/physiology , Receptors, Peptide/physiology , Uterus/physiology , Animals , Embryonic Development/genetics , Female , Fibroblasts/metabolism , Gene Deletion , Mice , Myeloid Cells/metabolism , Parturition/genetics , Pregnancy , Receptors, Peptide/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...