Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 35
1.
J Pharm Sci ; 105(12): 3507-3519, 2016 12.
Article En | MEDLINE | ID: mdl-27773526

Fibroblast growth factor-1 (FGF-1), a potent human mitogen and insulin sensitizer, signals through both tyrosine kinase receptor-mediated autocrine/paracrine pathways as well as a nuclear intracrine pathway. Phosphorylation of FGF-1 at serine 116 (S116) has been proposed to regulate intracrine signaling. Position S116 is located within a ∼17 amino acid C-terminal loop that contains a rich set of functional determinants including heparin∖heparan sulfate affinity, thiol reactivity, nuclear localization, pharmacokinetics, functional half-life, nuclear ligand affinity, stability, and structural dynamics. Mutational targeting of specific functionality in this region without perturbing other functional determinants is a design challenge. S116R is a non-phosphorylatable variant present in bovine FGF-1 and other members of the human FGF family. We show that the S116R mutation in human FGF-1 is accommodated with no perturbation of biophysical or structural properties, and is therefore an attractive mutation with which to elucidate the functional role of phosphorylation. Characterization of S116R shows reduction in NIH 3T3 fibroblast mitogenic stimulation, increase in fibroblast growth factor receptor-1c activation, and prolonged duration of glucose lowering in ob/ob hyperglycemic mice. A novel FGF-1/fibroblast growth factor receptor-1c dimerization interaction combined with non-phosphorylatable intracrine signaling is hypothesized to be responsible for these observed functional effects.


Fibroblast Growth Factor 1/genetics , Fibroblast Growth Factor 1/metabolism , Glucose/metabolism , Mitogens/metabolism , Mutation/physiology , Amino Acid Sequence , Animals , Cattle , Cell Survival/physiology , Crystallography, X-Ray , Dose-Response Relationship, Drug , Fibroblast Growth Factor 1/chemistry , Humans , Mice , NIH 3T3 Cells , Phosphorylation/physiology , Protein Structure, Secondary
2.
J Pharm Sci ; 105(4): 1444-53, 2016 Apr.
Article En | MEDLINE | ID: mdl-27019961

Human fibroblast growth factor-1 (FGF-1) has broad therapeutic potential in regenerative medicine but has undesirable biophysical properties of low thermostability and 3 buried cysteine (Cys) residues (at positions 16, 83, and 117) that interact to promote irreversible protein unfolding under oxidizing conditions. Mutational substitution of such Cys residues eliminates reactive buried thiols but cannot be accomplished simultaneously at all 3 positions without also introducing further substantial instability. The mutational introduction of a novel Cys residue (Ala66Cys) that forms a stabilizing disulfide bond (i.e., cystine) with one of the extant Cys residues (Cys83) effectively eliminates one Cys while increasing overall stability. This increase in stability offsets the associated instability of remaining Cys substitution mutations and permits production of a Cys-free form of FGF-1 (Cys16Ser/Ala66Cys/Cys117Ala) with only minor overall instability. The addition of a further stabilizing mutation (Pro134Ala) creates a Cys-free FGF-1 mutant with essentially wild-type biophysical properties. The elimination of buried free thiols in FGF-1 can substantially increase the protein half-life in cell culture. Here, we show that the effective cell survival/mitogenic functional activity of a fully Cys-free form is also substantially increased and is equivalent to wild-type FGF-1 formulated in the presence of heparin sulfate as a stabilizing agent. The results identify this Cys-free FGF-1 mutant as an advantageous "second generation" form of FGF-1 for therapeutic application.


Cysteine/chemistry , Fibroblast Growth Factor 1/chemistry , Amino Acid Substitution , Crystallography, X-Ray , Cysteine/genetics , Fibroblast Growth Factor 1/genetics , Humans , Models, Molecular , Protein Engineering , Protein Stability
3.
Wound Repair Regen ; 23(4): 538-49, 2015.
Article En | MEDLINE | ID: mdl-25891187

The development of novel therapies to treat chronic diabetic ulcers depends upon appropriate animal models for early stage investigation. The NONcNZO10/LtJ mouse is a new polygenic strain developed to more realistically model human metabolic syndrome and obesity-induced type 2 diabetes; however, detailed wound healing properties have not been reported. Herein, we describe a quantitative wound healing study in the NONcNZO10/LtJ mouse using a splinted excisional wound. The rate of wound healing is compared to various controls, and is also quantified in response to topical administration of normal and mutant fibroblast growth factor-1 (FGF-1). Quantitation of reepithelialization shows that the diabetic condition in the NONcNZO10/LtJ mouse is concomitant with a decreased rate of dermal healing. Furthermore, topical administration of a FGF-1/heparin formulation effectively accelerates reepithelialization. A similar acceleration can also be achieved by a stabilized mutant form of FGF-1 formulated in the absence of heparin. Such accelerated rates of healing are not associated with any abnormal histology in the healed wounds. The results identify the NONcNZO10/LtJ mouse as a useful model of impaired wound healing in type 2 diabetes, and further, identify engineered forms of FGF-1 as a potential "second-generation" therapeutic to promote diabetic dermal wound healing.


Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2/complications , Fibroblast Growth Factor 1/administration & dosage , Heparin/administration & dosage , Mutant Proteins/administration & dosage , Skin Ulcer/pathology , Wound Healing/physiology , Administration, Topical , Animals , Chronic Disease , Diabetes Mellitus, Type 2/pathology , Drug Combinations , Male , Mice , Re-Epithelialization/drug effects , Skin/drug effects , Skin/pathology , Skin Ulcer/etiology , Treatment Outcome , Wound Healing/drug effects
4.
Brain Inj ; 27(13-14): 1698-706, 2013.
Article En | MEDLINE | ID: mdl-24266798

BACKGROUND: Establishment of a traumatic brain injury (TBI)-sensitive biomarker or identification of a key therapeutic agent would significantly improve clinicians' efforts to diagnose and treat TBI, thereby promoting improved outcomes for patients. Numerous studies support the role of kallikrein-6 (Klk6) as a critical component of neuroinflammation and demyelination. This study assesses whether Klk6 is implicated in the secondary mechanisms of TBI and subsequently if serum levels of Klk6 are useable as a biomarker. METHODS: The abundance of Klk6 following controlled cortical impact (CCI) of the medial prefrontal cortex to a depth of either 3.0 mm (severe) or 1.5 mm (moderate) was quantified. Uninjured and rats subjected to craniotomy-only were used as controls. Protein levels were quantified with Western-blotting, enzyme-linked immunosorbent assay and immunohistochemistry. RESULTS: Severe and moderate CCI resulted in significant elevation of Klk6 in the contusion-core (~12-fold-increase, p < 0.0001) and serum (~5-fold-increase, p < 0.01) compared to controls. In all cases, Klk6 elevation was resolved within 72 hours. CONCLUSION: Serum levels of Klk6 are a statistically significant indicator of TBI 24 hours after CCI and thus may be of great utility to clinicians as a biomarker. These data strongly implicate Klk6 as a player in the neuroinflammation processes following CCI, although the specific mechanisms remain to be characterized.


Brain Injuries/metabolism , Demyelinating Diseases/metabolism , Inflammation/metabolism , Kallikreins/metabolism , Animals , Biomarkers/metabolism , Blotting, Western , Brain Injuries/complications , Demyelinating Diseases/etiology , Immunohistochemistry , Inflammation/etiology , Male , Rats , Rats, Sprague-Dawley , Trauma Severity Indices
5.
J Neuropathol Exp Neurol ; 72(11): 1072-89, 2013 Nov.
Article En | MEDLINE | ID: mdl-24128681

Kallikreins (KLKs) are a family of 15 secreted serine proteases with emerging roles in neurologic diseases. To illuminate their contributions to the pathophysiology of spinal cord injury (SCI), we evaluated acute through chronic changes in the immunohistochemical appearance of 6 KLKs (KLK1, KLK5, KLK6, KLK7, KLK8, and KLK9) in postmortem human traumatic SCI cases, quantified their RNA expression levels in experimental murine SCI, and assessed the impact of recombinant forms of each enzyme toward murine cortical neurons in vitro. Temporally and spatially distinct changes in KLK expression were observed with partially overlapping patterns between human and murine SCI, including peak elevations (or reductions) during the acute and subacute periods. Kallikrein 9 showed the most marked changes and remained chronically elevated. Importantly, a subset of KLKs (KLK1, KLK5, KLK6, KLK7, and KLK9) were neurotoxic toward primary neurons in vitro. Kallikrein immunoreactivity was also observed in association with swollen axons and retraction bulbs in the human SCI cases examined. Together, these findings demonstrate that elevated levels of a significant subset of KLKs are positioned to contribute to neurodegenerative changes in cases of CNS trauma and disease and, therefore, represent new potential targets for the development of neuroprotective strategies.


Axons/metabolism , Kallikreins/metabolism , Nerve Degeneration/metabolism , Spinal Cord Injuries/metabolism , Spinal Cord/metabolism , Adolescent , Adult , Aged , Animals , Axons/pathology , Child , Child, Preschool , Female , Humans , Kallikreins/genetics , Male , Mice , Middle Aged , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Signal Transduction/physiology , Spinal Cord/pathology , Spinal Cord Injuries/genetics , Spinal Cord Injuries/pathology
6.
J Neurochem ; 127(2): 283-98, 2013 Oct.
Article En | MEDLINE | ID: mdl-23647384

CNS trauma generates a proteolytic imbalance contributing to secondary injury, including axonopathy and neuron degeneration. Kallikrein 6 (Klk6) is a serine protease implicated in neurodegeneration, and here we investigate the role of protease-activated receptors 1 (PAR1) and PAR2 in mediating these effects. First, we demonstrate Klk6 and the prototypical activator of PAR1, thrombin, as well as PAR1 and PAR2, are each elevated in murine experimental traumatic spinal cord injury (SCI) at acute or subacute time points. Recombinant Klk6 triggered extracellular signal-regulated kinase (ERK1/2) signaling in cerebellar granule neurons and in the NSC34 spinal cord motoneuron cell line, in a phosphoinositide 3-kinae and MEK-dependent fashion. Importantly, lipopeptide inhibitors of PAR1 or PAR2, and PAR1 genetic deletion, each reduced Klk6-ERK1/2 activation. In addition, Klk6 and thrombin promoted degeneration of cerebellar neurons and exacerbated glutamate neurotoxicity. Moreover, genetic deletion of PAR1 blocked thrombin-mediated cerebellar neurotoxicity and reduced the neurotoxic effects of Klk6. Klk6 also increased glutamate-mediated Bim signaling, poly-ADP-ribose polymerase cleavage and lactate dehydrogenase release in NSC34 motoneurons and these effects were blocked by PAR1 and PAR2 lipopeptide inhibitors. Taken together, these data point to a novel Klk6-signaling axis in CNS neurons that is mediated by PAR1 and PAR2 and is positioned to contribute to neurodegeneration.


Glutamic Acid/toxicity , Kallikreins/physiology , Neurons/pathology , Neurotoxicity Syndromes/physiopathology , Receptor, PAR-1/physiology , Receptor, PAR-2/physiology , Animals , Cells, Cultured , Cerebellum/cytology , Female , Kallikreins/genetics , MAP Kinase Signaling System/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Neurons/physiology , RNA/biosynthesis , RNA/genetics , Receptor, PAR-1/genetics , Receptor, PAR-2/genetics , Signal Transduction/physiology , Spinal Cord/cytology , Spinal Cord/physiology , Spinal Cord Injuries/pathology , Thrombin/genetics , Thrombin/physiology
7.
Neuro Oncol ; 15(3): 305-18, 2013 Mar.
Article En | MEDLINE | ID: mdl-23307575

BACKGROUND: Kallikreins have prognostic value in specific malignancies, but few studies have addressed their clinical significance to glioblastoma multiforme (GBM). Kallikrein 6 (KLK6) is of potential high relevance to GBM, since it is upregulated at sites of CNS pathology and linked to reactive astrogliosis. Here we examine the clinical value of KLK6 as a prognostic indicator of GBM patient survival and its activity in promoting resistance to cytotoxic agents. METHODS: The association between patient survival and levels of KLK6 immunoreactivity were investigated in 60 grade IV astrocytoma tumor specimens. Levels of KLK6 RNA were also evaluated in a separate set of GBM patient tumors (n = 23). Recombinant KLK6 or enforced KLK6 overexpression in GBM cell lines was used to evaluate effects on astrocytoma cell survival. RESULTS: A range of KLK6 expression was observed across grade IV tumors, with higher levels a poor prognostic indicator of patient survival (P = .02) even after adjusting for gender and Eastern Cooperative Oncology Group performance scores (P = .01). KLK6 reduced the sensitivity of GBM cell lines to cytotoxic agents, including staurosporine and cisplatin, and to the current standard of patient care: radiotherapy or temozolomide alone or in combination. The ability of KLK6 to promote resistance to apoptosis was dependent on activation of the thrombin receptor, protease activated receptor 1. CONCLUSIONS: Taken together, these results indicate that elevated levels of KLK6 in GBM are likely to promote the resistance of tumor cells to cytotoxic agents and are an indicator of reduced patient postsurgical survival times.


Apoptosis/drug effects , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Kallikreins/metabolism , Recombinant Proteins/metabolism , Antineoplastic Agents/pharmacology , Blotting, Western , Brain Neoplasms/drug therapy , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Cell Proliferation/drug effects , Cisplatin/pharmacology , Enzyme Inhibitors/pharmacology , Glioblastoma/drug therapy , Glioblastoma/mortality , Glioblastoma/pathology , Humans , Kallikreins/genetics , Neoplasm Grading , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Recombinant Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Staurosporine/pharmacology , Survival Rate , Tumor Cells, Cultured
8.
Biol Chem ; 394(1): 137-47, 2013 Jan.
Article En | MEDLINE | ID: mdl-23241590

The 15 human kallikrein-related peptidases (KLKs) are clinically important biomarkers and therapeutic targets of interest in inflammation, cancer, and neurodegenerative disease. KLKs are secreted as inactive pro-forms (pro-KLKs) that are activated extracellularly by specific proteolytic release of their amino-terminal pro-peptide, and this is a key step in their functional regulation. Physiologically relevant KLK regulatory cascades of activation have been described in skin desquamation and semen liquefaction, and work by a large number of investigators has elucidated pairwise and autolytic activation relationships among the KLKs with the potential for more extensive activation cascades. More recent work has asked whether functional intersection of KLKs with other types of regulatory proteases exists. Such studies show a capacity for members of the thrombostasis axis to act as broad activators of pro-KLKs. In the present report, we ask whether such functional intersection is possible between the KLKs and the members of the matrix metalloproteinase (MMP) family by evaluating the ability of the MMPs to activate pro-KLKs. The results identify MMP-20 as a broad activator of pro-KLKs, suggesting the potential for intersection of the KLK and MMP axes under pathological dysregulation of MMP-20 expression.


Kallikreins/metabolism , Matrix Metalloproteinase 20/metabolism , Enzyme Activation , Humans , Hydrolysis
9.
PLoS One ; 7(11): e48210, 2012.
Article En | MEDLINE | ID: mdl-23133616

Fibroblast growth factor-1 (FGF-1) is an angiogenic factor with therapeutic potential for the treatment of ischemic disease. FGF-1 has low intrinsic thermostability and is characteristically formulated with heparin as a stabilizing agent. Heparin, however, adds a number of undesirable properties that negatively impact safety and cost. Mutations that increase the thermostability of FGF-1 may obviate the need for heparin in formulation and may prove to be useful "2nd-generation" forms for therapeutic use. We report a pharmacokinetic (PK) study in rabbits of human FGF-1 in the presence and absence of heparin, as well as three mutant forms having differential effects upon thermostability, buried reactive thiols, and heparin affinity. The results support the hypothesis that heparan sulfate proteoglycan (HSPG) in the vasculature of liver, kidney and spleen serves as the principle peripheral compartment in the distribution kinetics. The addition of heparin to FGF-1 is shown to increase endocrine-like properties of distribution. Mutant forms of FGF-1 that enhance thermostability or eliminate buried reactive thiols demonstrate a shorter distribution half-life, a longer elimination half-life, and a longer mean residence time (MRT) in comparison to wild-type FGF-1. The results show how such mutations can produce useful 2nd-generation forms with tailored PK profiles for specific therapeutic application.


Fibroblast Growth Factor 1/genetics , Fibroblast Growth Factor 1/pharmacokinetics , Ischemia/drug therapy , Mutation , Animals , Blood Glucose/metabolism , Escherichia coli/metabolism , Heparan Sulfate Proteoglycans/pharmacokinetics , Humans , Kinetics , Male , Protein Conformation , Rabbits , Recombinant Proteins/chemistry , Sulfhydryl Compounds/chemistry , Triglycerides/metabolism
10.
Biol Chem ; 393(5): 355-67, 2012 Apr.
Article En | MEDLINE | ID: mdl-22505518

Kallikrein-related peptidase 6 (KLK6) is a trypsin-like serine protease upregulated at sites of central nervous system (CNS) injury, including de novo expression by reactive astrocytes, yet its physiological actions are largely undefined. Taken with recent evidence that KLK6 activates G-protein-coupled protease-activated receptors (PARs), we hypothesized that injury-induced elevations in KLK6 contribute to the development of astrogliosis and that this occurs in a PAR-dependent fashion. Using primary murine astrocytes and the Neu7 astrocyte cell line, we show that KLK6 causes astrocytes to transform from an epitheliod to a stellate morphology and to secrete interleukin 6 (IL-6). By contrast, KLK6 reduced expression of glial fibrillary acidic protein (GFAP). The stellation-promoting activities of KLK6 were shown to be dependent on activation of the thrombin receptor, PAR1, as a PAR1-specific inhibitor, SCH79797, blocked KLK6-induced morphological changes. The ability of KLK6 to promote astrocyte stellation was also shown to be linked to activation of protein kinase C (PKC). These studies indicate that KLK6 is positioned to serve as a molecular trigger of select physiological processes involved in the development of astrogliosis and that this is likely to occur at least in part by activation of the G-protein-coupled receptor, PAR1.


Gliosis/enzymology , Gliosis/pathology , Kallikreins/metabolism , Animals , Astrocytes/enzymology , Astrocytes/metabolism , Astrocytes/pathology , Brain/enzymology , Brain/metabolism , Brain/pathology , Carrier Proteins/metabolism , Cell Proliferation , DNA-Binding Proteins , Gene Expression Regulation , Glial Fibrillary Acidic Protein/metabolism , Gliosis/metabolism , Humans , Hypertrophy , Interleukin-6/metabolism , Mice , Protein Kinase C/metabolism , Receptor, PAR-1/metabolism , Spinal Cord/enzymology , Spinal Cord/metabolism , Spinal Cord/pathology
11.
Brain Pathol ; 22(5): 709-22, 2012 Sep.
Article En | MEDLINE | ID: mdl-22335454

Kallikrein 6 (Klk6) is a secreted serine protease that is elevated in active multiple sclerosis lesions and patient sera. To further evaluate the involvement of Klk6 in chronic progressive demyelinating disease, we determined its expression in the brain and spinal cord of SJL mice infected with Theiler's murine encephalomyelitis virus (TMEV) and assessed the effects of Klk6-neutralizing antibodies on disease progression. Klk6 RNA expression was elevated in the brain and spinal cord by 7 days postinfection (dpi). Thereafter, Klk6 expression persisted primarily in the spinal cord reaching a peak of fivefold over controls at mid-chronic stages (60 dpi-120 dpi). Significant elevations in Klk6 RNA were also induced in splenocytes stimulated with viral capsid proteins in vitro and in activated human acute monocytic leukemia cells. Klk6-neutralizing antibodies reduced TMEV-driven brain and spinal cord pathology and delayed-type hypersensitivity (DTH) responses when examined at early chronic time points (40 dpi). Reductions in spinal cord pathology included a decrease in activated monocytes/microglia and reductions in the loss of myelin basic protein (MBP). By 180 dpi, pathology scores no longer differed between groups. These findings point to regulatory activities for Klk6 in the development and progression of central nervous system (CNS) inflammation and demyelination that can be effectively targeted through the early chronic stages with neutralizing antibody.


Central Nervous System/metabolism , Kallikreins/metabolism , Multiple Sclerosis/etiology , Multiple Sclerosis/pathology , Multiple Sclerosis/virology , Theilovirus/pathogenicity , Analysis of Variance , Animals , Antibodies/therapeutic use , Disease Models, Animal , Gene Expression Regulation, Viral/drug effects , Hypersensitivity/etiology , Kallikreins/genetics , Kallikreins/immunology , Mice , Monocytes/metabolism , Multiple Sclerosis/therapy , RNA, Messenger/metabolism , RNA, Viral/genetics , Spinal Cord/metabolism , Statistics, Nonparametric , T-Lymphocytes/metabolism , Theilovirus/genetics , Time Factors , Up-Regulation/genetics
12.
J Invest Dermatol ; 131(11): 2281-8, 2011 Nov.
Article En | MEDLINE | ID: mdl-21753781

Cutaneous malignant melanoma is an aggressive disease of poor prognosis. Clinical and experimental studies have provided major insight into the pathogenesis of the disease, including the functional interaction between melanoma cells and surrounding keratinocytes, fibroblasts, and immune cells. Nevertheless, patients with metastasized melanoma have a very poor prognosis and are largely refractory to clinical therapies. Hence, diagnostic tools to monitor melanoma development, as well as therapeutic targets, are urgently needed. We investigated the expression pattern of the kallikrein-related peptidase 6 (KLK6) in human melanoma tissue sections throughout tumor development. Although KLK6 was not detectable in tumor cells, we found strong KLK6 protein expression in keratinocytes and stromal cells located adjacent to benign nevi, primary melanomas, and cutaneous metastatic lesions, suggesting a paracrine function of extracellular KLK6 during neoplastic transformation and malignant progression. Accordingly, recombinant Klk6 protein significantly induced melanoma cell migration and invasion accompanied by an accelerated intracellular Ca(2+) flux. We could further demonstrate that KLK6-induced intracellular Ca(2+) flux and tumor cell invasion critically depends on the protease-activated receptor 1 (PAR1). Our data provide experimental evidence that specific inhibition of the KLK6-PAR1 axis may interfere with the deleterious effect of tumor-microenvironment interaction and represent a potential option for translational melanoma research.


Kallikreins/metabolism , Melanoma/metabolism , Melanoma/physiopathology , Skin Neoplasms/metabolism , Skin Neoplasms/physiopathology , Tumor Microenvironment/physiology , Aged , Aged, 80 and over , Biopsy , Calcium/metabolism , Cell Line, Tumor , Cell Movement/physiology , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cells, Cultured , Female , Humans , Male , Melanoma/pathology , Middle Aged , Receptor, PAR-1/metabolism , Signal Transduction/physiology , Skin/pathology , Skin Neoplasms/pathology
13.
Biochimie ; 93(10): 1701-9, 2011 Oct.
Article En | MEDLINE | ID: mdl-21689719

KLK13 is a kallikrein-related peptidase preferentially expressed in tonsils, esophagus, testis, salivary glands and cervix. We report the activation of KLK13 by kosmotropic salts and glycosaminoglycans and its substrate specificity by employing a series of five substrates derived from the fluorescence resonance energy transfer (FRET) peptide Abz-KLRSSKQ-EDDnp. KLK13 hydrolyzed all these peptides only at basic residues with highest efficiency for R; furthermore, the S(3) to S(2)' subsites accepted most of the natural amino acids with preference also for basic residues. Using a support-bound FRET peptide library eight peptide substrates were identified containing sequences of proteins found in testis and one with myelin basic protein sequence, each of which was well hydrolyzed by KLK13. Histatins are salivary peptides present in higher primates with broad antifungal and mucosal healing activities that are generated from the hydrolysis from large precursor peptides. KLK13 efficiently hydrolyzed synthetic histatin 3 exclusively at R(25) (DSHAKRHHGYKRKFHEKHHSHRGYR(25)↓SNYLYDN) that is the first cleavage observed inside the salivary gland. In conclusion, the observed hydrolytic activities of KLK13 and its co-localization with its activators, glycosaminoglycans in the salivary gland and high concentration of sodium citrate in male reproductive tissues, indicates that KLK13 may play a role in the defense of the upper digestive apparatus and in male reproductive organs.


Glycosaminoglycans/pharmacology , Kallikreins/metabolism , Salts/pharmacology , Citrates/pharmacology , Enzyme Activation/drug effects , Female , Humans , Male , Salivary Glands/metabolism , Sodium Citrate , Substrate Specificity
14.
PLoS One ; 6(3): e18376, 2011 Mar 28.
Article En | MEDLINE | ID: mdl-21464892

BACKGROUND: Kallikrein 6 (KLK6) is a newly identified member of the kallikrein family of secreted serine proteases that prior studies indicate is elevated at sites of central nervous system (CNS) inflammation and which shows regulated expression with T cell activation. Notably, KLK6 is also elevated in the serum of multiple sclerosis (MS) patients however its potential roles in immune function are unknown. Herein we specifically examine whether KLK6 alters immune cell survival and the possible mechanism by which this may occur. METHODOLOGY/PRINCIPAL FINDINGS: Using murine whole splenocyte preparations and the human Jurkat T cell line we demonstrate that KLK6 robustly supports cell survival across a range of cell death paradigms. Recombinant KLK6 was shown to significantly reduce cell death under resting conditions and in response to camptothecin, dexamethasone, staurosporine and Fas-ligand. Moreover, KLK6-over expression in Jurkat T cells was shown to generate parallel pro-survival effects. In mixed splenocyte populations the vigorous immune cell survival promoting effects of KLK6 were shown to include both T and B lymphocytes, to occur with as little as 5 minutes of treatment, and to involve up regulation of the pro-survival protein B-cell lymphoma-extra large (Bcl-XL), and inhibition of the pro-apoptotic protein Bcl-2-interacting mediator of cell death (Bim). The ability of KLK6 to promote survival of splenic T cells was also shown to be absent in cell preparations derived from PAR1 deficient mice. CONCLUSION/SIGNIFICANCE: KLK6 promotes lymphocyte survival by a mechanism that depends in part on activation of PAR1. These findings point to a novel molecular mechanism regulating lymphocyte survival that is likely to have relevance to a range of immunological responses that depend on apoptosis for immune clearance and maintenance of homeostasis.


Kallikreins/pharmacology , Lymphocytes/cytology , Lymphocytes/drug effects , Animals , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Cell Death/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Jurkat Cells , Lymphocytes/metabolism , Mice , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptor, PAR-1/metabolism , Signal Transduction/drug effects , Spleen/cytology , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
15.
J Mol Biol ; 407(5): 744-63, 2011 Apr 15.
Article En | MEDLINE | ID: mdl-21315087

Fibroblast growth factor-1, a member of the 3-fold symmetric ß-trefoil fold, was subjected to a series of symmetric constraint mutations in a process termed "top-down symmetric deconstruction." The mutations enforced a cumulative exact 3-fold symmetry upon symmetrically equivalent positions within the protein and were combined with a stability screen. This process culminated in a ß-trefoil protein with exact 3-fold primary-structure symmetry that exhibited excellent folding and stability properties. Subsequent fragmentation of the repeating primary-structure motif yielded a 42-residue polypeptide capable of spontaneous assembly as a homotrimer, producing a thermostable ß-trefoil architecture. The results show that despite pronounced reduction in sequence complexity, pure symmetry in the design of a foldable, thermostable ß-trefoil fold is possible. The top-down symmetric deconstruction approach provides a novel alternative means to successfully identify a useful polypeptide "building block" for subsequent "bottom-up" de novo design of target protein architecture.


Peptides/chemistry , Protein Folding , Protein Structure, Secondary , Protein Structure, Tertiary , 3T3 Cells , Amino Acid Sequence , Animals , Crystallography, X-Ray , Fibroblast Growth Factor 1/chemistry , Fibroblast Growth Factor 1/genetics , Humans , Mice , Molecular Sequence Data , Peptides/genetics , Protein Denaturation , Receptor, Fibroblast Growth Factor, Type 1/chemistry , Receptor, Fibroblast Growth Factor, Type 1/genetics , Sequence Alignment , Sequence Homology, Amino Acid
16.
Arch Biochem Biophys ; 498(1): 74-82, 2010 Jun 01.
Article En | MEDLINE | ID: mdl-20371359

We report the enzymatic properties and substrate specificity of human recombinant KLK3 in the presence of glycosaminoglycans (GAGs) and sodium citrate. This salt is highly concentrated in prostate and in its presence KLK3 had a similar hydrolytic efficiency as chymotrypsin. In contrast to the latter peptidase, KLK3 activated by sodium citrate efficiently hydrolyzed substrates containing R, H and P at the P1 position. Activated KLK3 also cleaved peptides derived from the bradykinin domain of human kininogen at the same sites as human kallikrein KLK1, but presented low kininogenase activity. Angiotensin I has several sites for hydrolysis by KLK3; however, it was cleaved only at the Y-I bond (DRVY downward arrowIHPFHL). Sodium citrate modulated KLK3 conformation as observed by alterations to the intrinsic fluorescence of phenylalanines and tryptophans. Activated KLK3 was reversibly inhibited by Z-Pro-Prolinal and competitively inhibited by ortho-phenantroline. Together, these are noteworthy observations for the future design of specific non-peptide inhibitors of KLK3 and to find natural substrates.


Citrates/pharmacology , Glycosaminoglycans/pharmacology , Kallikreins/antagonists & inhibitors , Kallikreins/metabolism , Protease Inhibitors/pharmacology , Amino Acid Sequence , Angiotensins/metabolism , Chlorides/chemistry , Chlorides/pharmacology , Enzyme Activation/drug effects , Fluorescence Resonance Energy Transfer , Humans , Hydrogen-Ion Concentration , Hydrolysis , Kallikreins/chemistry , Kininogen, Low-Molecular-Weight/metabolism , Molecular Sequence Data , Prostate-Specific Antigen , Salts/chemistry , Salts/pharmacology , Sodium Citrate , Substrate Specificity
17.
Biol Chem ; 391(4): 311-20, 2010 Apr.
Article En | MEDLINE | ID: mdl-20128685

A large body of emerging evidence indicates a functional interaction between the kallikrein-related peptidases (KLKs) and proteases of the thrombostasis axis. These interactions appear relevant for both normal health as well as pathologies associated with inflammation, tissue injury, and remodeling. Regulatory interactions between the KLKs and thrombostasis proteases could impact several serious human diseases, including neurodegeneration and cancer. The emerging network of specific interactions between these two protease families appears to be complex, and much work remains to elucidate it. Complete understanding how this functional network resolves over time, given specific initial conditions, and how it might be controllably manipulated, will probably contribute to the emergence of novel diagnostics and therapeutic agents for major diseases.


Kallikreins/metabolism , Thrombosis/metabolism , Angiostatins/chemistry , Angiostatins/metabolism , Animals , Enzyme Activation , Humans , Protease Inhibitors/metabolism , Receptors, Proteinase-Activated/metabolism
18.
Acta Crystallogr Sect F Struct Biol Cryst Commun ; 65(Pt 11): 1097-104, 2009 Nov 01.
Article En | MEDLINE | ID: mdl-19923726

The rabbit is an important and de facto animal model in the study of ischemic disease and angiogenic therapy. Additionally, fibroblast growth factor 1 (FGF-1) is emerging as one of the most important growth factors for novel proangiogenic and pro-arteriogenic therapy. However, despite its significance, the fundamental biophysical properties of rabbit FGF-1, including its X-ray structure, have never been reported. Here, the cloning, crystallization, X-ray structure and determination of the biophysical properties of rabbit FGF-1 are described. The X-ray structure shows that the amino-acid differences between human and rabbit FGF-1 are solvent-exposed and therefore potentially immunogenic, while the biophysical studies identify differences in thermostability and receptor-binding affinity that distinguish rabbit FGF-1 from human FGF-1.


Fibroblast Growth Factor 1/chemistry , Protein Structure, Secondary , Protein Structure, Tertiary , Amino Acid Sequence , Animals , Crystallography, X-Ray , Female , Humans , Models, Molecular , Molecular Sequence Data , Rabbits , Sequence Alignment , Sequence Homology, Amino Acid , Thermodynamics
19.
Acta Crystallogr Sect F Struct Biol Cryst Commun ; 65(Pt 11): 1136-40, 2009 Nov 01.
Article En | MEDLINE | ID: mdl-19923735

Large-volume protein crystals are a prerequisite for neutron diffraction studies and their production represents a bottleneck in obtaining neutron structures. Many protein crystals that permit the collection of high-resolution X-ray diffraction data are inappropriate for neutron diffraction owing to a plate-type morphology that limits the crystal volume. Human fibroblast growth factor 1 crystallizes in a plate morphology that yields atomic resolution X-ray diffraction data but has insufficient volume for neutron diffraction. The thin physical dimension has been identified as corresponding to the b cell edge and the X-ray structure identified a solvent-mediated crystal contact adjacent to position Glu81 that was hypothesized to limit efficient crystal growth in this dimension. In this report, a series of mutations at this crystal contact designed to both reduce side-chain entropy and replace the solvent-mediated interface with direct side-chain contacts are reported. The results suggest that improved crystal growth is achieved upon the introduction of direct crystal contacts, while little improvement is observed with side-chain entropy-reducing mutations alone.


Crystallization/methods , Fibroblast Growth Factor 1/chemistry , Protein Engineering/methods , Solvents/chemistry , Crystallography, X-Ray , Fibroblast Growth Factor 1/genetics , Humans , Models, Molecular , Molecular Sequence Data , Mutagenesis , Neutron Diffraction , Protein Conformation , X-Ray Diffraction
20.
Biol Chem ; 390(4): 373-7, 2009 Apr.
Article En | MEDLINE | ID: mdl-19090718

We previously reported the activation profiles of the human kallikrein-related peptidases (KLKs) as determined from a KLK pro-peptide fusion-protein system. That report described the activity profiles of 12 of the 15 mature KLKs versus the 15 different pro-KLK sequences. The missing profiles in the prior report, involving KLK9, 10, and 15, are now described. Sodium dodecyl sulfate-polyacrylamide gel electrophoresis, mass spectrometry, and N-terminal sequence analyses show that KLK9 and 10 exhibit low hydrolytic activities towards all of the 15 pro-KLK sequences, while KLK15 exhibits significant activity towards both Arg- and Lys-containing KLK pro-sequences. The ability of KLK15 to activate pro-KLK8, 12, and 14 is confirmed using recombinant pro-KLK proteins, and shown to be significant for activation of pro-KLK8 and 14, but not 12. These additional data for KLK9, 10, and 15 now permit a completed KLK activome profile, using a KLK pro-peptide fusion-protein system, to be described. The results suggest that KLK15, once activated, can potentially feed back into additional pro-KLK activation pathways. Conversely, KLK9 and 10, once activated, are unlikely to participate in further pro-KLK activation pathways, although similar to KLK1 they may activate other bioactive peptides.


Kallikreins/metabolism , Electrophoresis, Agar Gel , Gene Expression Profiling , Humans , Kallikreins/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
...