Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 83
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Med Microbiol Immunol ; 213(1): 9, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38900248

ABSTRACT

Endogenous antimicrobial peptides (AMPs) play a key role in the host defense against pathogens. AMPs attack pathogens preferentially at the site of entry to prevent invasive infection. Mycobacterium tuberculosis (Mtb) enters its host via the airways. AMPs released into the airways are therefore likely candidates to contribute to the clearance of Mtb immediately after infection. Since lysozyme is detectable in airway secretions, we evaluated its antimicrobial activity against Mtb. We demonstrate that lysozyme inhibits the growth of extracellular Mtb, including isoniazid-resistant strains. Lysozyme also inhibited the growth of non-tuberculous mycobacteria. Even though lysozyme entered Mtb-infected human macrophages and co-localized with the pathogen we did not observe antimicrobial activity. This observation was unlikely related to the large size of lysozyme (14.74 kDa) because a smaller lysozyme-derived peptide also co-localized with Mtb without affecting the viability. To evaluate whether the activity of lysozyme against extracellular Mtb could be relevant in vivo, we incubated Mtb with fractions of human serum and screened for antimicrobial activity. After several rounds of sub-fractionation, we identified a highly active fraction-component as lysozyme by mass spectrometry. In summary, our results identify lysozyme as an antimycobacterial protein that is detectable as an active compound in human serum. Our results demonstrate that the activity of AMPs against extracellular bacilli does not predict efficacy against intracellular pathogens despite co-localization within the macrophage. Ongoing experiments are designed to unravel peptide modifications that occur in the intracellular space and interfere with the deleterious activity of lysozyme in the extracellular environment.


Subject(s)
Macrophages , Muramidase , Mycobacterium tuberculosis , Muramidase/pharmacology , Muramidase/metabolism , Humans , Macrophages/metabolism , Macrophages/microbiology , Antimicrobial Peptides/pharmacology , Antimicrobial Peptides/metabolism , Microbial Sensitivity Tests , Microbial Viability/drug effects
2.
Proc Natl Acad Sci U S A ; 118(3)2021 01 19.
Article in English | MEDLINE | ID: mdl-33431697

ABSTRACT

GPR15 is a G protein-coupled receptor (GPCR) proposed to play a role in mucosal immunity that also serves as a major entry cofactor for HIV-2 and simian immunodeficiency virus (SIV). To discover novel endogenous GPR15 ligands, we screened a hemofiltrate (HF)-derived peptide library for inhibitors of GPR15-mediated SIV infection. Our approach identified a C-terminal fragment of cystatin C (CysC95-146) that specifically inhibits GPR15-dependent HIV-1, HIV-2, and SIV infection. In contrast, GPR15L, the chemokine ligand of GPR15, failed to inhibit virus infection. We found that cystatin C fragments preventing GPR15-mediated viral entry do not interfere with GPR15L signaling and are generated by proteases activated at sites of inflammation. The antiretroviral activity of CysC95-146 was confirmed in primary CD4+ T cells and is conserved in simian hosts of SIV infection. Thus, we identified a potent endogenous inhibitor of GPR15-mediated HIV and SIV infection that does not interfere with the physiological function of this GPCR.


Subject(s)
Cystatin C/genetics , HIV Infections/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, Peptide/genetics , Simian Acquired Immunodeficiency Syndrome/genetics , Animals , HIV Infections/pathology , HIV Infections/virology , HIV-1/genetics , HIV-1/pathogenicity , Humans , Receptors, Virus/genetics , Signal Transduction/genetics , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/pathogenicity , T-Lymphocytes/metabolism , T-Lymphocytes/virology , Virus Internalization
3.
Eur J Immunol ; 48(9): 1592-1594, 2018 09.
Article in English | MEDLINE | ID: mdl-30028015

ABSTRACT

Chemokine CCL14 is inactive in its proform. Here, we show that inflammation- and cancer-associated kallikrein-related peptidases KLK5 and KLK8 remove the N-terminal eight amino acids from the proform thereby converting CCL14 to its active state. Activity of the chemokine is demonstrated by migration of myeloid cells expressing relevant receptors.


Subject(s)
Chemokines, CC/metabolism , Chemokines/metabolism , Kallikreins/metabolism , Asthma/pathology , Atherosclerosis/pathology , Cell Line, Tumor , Chemokine CX3CL1/metabolism , Chemokine CXCL12/metabolism , Crohn Disease/pathology , Enzyme Activation , Humans , Interleukin-8/metabolism , Leukemia/pathology , Macrophage Inflammatory Proteins/metabolism , Pancreatitis/pathology , Reactive Oxygen Species/metabolism
4.
Kidney Blood Press Res ; 41(5): 507-518, 2016.
Article in English | MEDLINE | ID: mdl-27487342

ABSTRACT

BACKGROUND/AIMS: Parathyroid hormone (PTH) derivatives exert pronounced renal and osteoanabolic properties when given intermittently. The current study was performed to assess the pharmacokinetic and pharmacodynamic properties as well as safety of subcutaneously applied PTH-1-37 after repeated dosing in healthy subjects. METHODS: This randomized, double-blind, dose-escalating, placebo and active comparator controlled study was conducted in 33 healthy postmenopausal women. Subjects were allocated to one of five treatment options: 10, 20, or 40 µg PTH-1-37, 20 µg PTH-1-34 or placebo, administered as once daily subcutaneous doses for three days. Plasma drug concentrations and serum levels of endogenous PTH-1-84, and calcium as markers of biological activity were monitored during the treatment. RESULTS: PTH was absorbed rapidly from the subcutaneous tissue with a median tmax of 30 minutes for 20 and 40 µg of PTH-1-37. tmax was 45 minutes for 20 µg PTH-1-34. Elimination half-lives were estimated as 76 ± 34 min and 70 ± 13 min for 20 µg and 40 µg PTH-1-37 (mean ± SD), and 78 ± 34 for 20 µg PTH-1-34. Both PTH fragments (PTH-1-37 and PTH-1-34) increased serum calcium. For PTH-1-37 the effect on serum calcium was dose-dependent. Suppression of endogenous PTH-1-84 was seen after the application of both PTH-1-37 and PTH-1-34. During the study period, the subjects experienced no unexpected or serious adverse events. CONCLUSIONS: PTH-1-37 is rapidly absorbed after s.c. injection, has a short plasma elimination half-life, and does not accumulate during multiple dosing. Biological activity was demonstrated by rising serum calcium and decreasing endogenous PTH-1-84 in blood plasma. The study drugs were well tolerated and safe. Our investigation presents data that PTH-1-37 is an excellent drug candidate for intervening with syndromes of dysregulation of calcium metabolism.


Subject(s)
Parathyroid Hormone/pharmacokinetics , Peptide Fragments/pharmacokinetics , Calcium/blood , Calcium/metabolism , Calcium Metabolism Disorders/drug therapy , Dose-Response Relationship, Drug , Double-Blind Method , Female , Half-Life , Healthy Volunteers , Humans , Injections, Subcutaneous , Middle Aged , Parathyroid Hormone/administration & dosage , Parathyroid Hormone/blood , Peptide Fragments/administration & dosage
5.
Kidney Blood Press Res ; 40(1): 77-88, 2015.
Article in English | MEDLINE | ID: mdl-25791819

ABSTRACT

BACKGROUND/AIMS: In diabetic nephropathy (DN), the current angiotensin-II-blocking pharmacotherapy is frequently failing. For diabetic cardiomyopathy (DC), there is no specific remedy available. Relaxin-2 (Rlx) - an anti-fibrotic, anti-inflammatory, and vasoprotecting peptide ­ is a candidate drug for both. METHODS: Low-dose (32 µg/kg/day) and high-dose (320 µg/kg/day) Rlx were tested against vehicle (n = 20 each) and non-diabetic controls (n = 14) for 12 weeks in a model of type-1 diabetes induced in endothelial nitric oxide synthase knock-out (eNOS-KO) mice by intraperitoneal injection of streptozotocin. RESULTS: Diabetic animals showed normal plasma creatinine, markedly increased albuminuria and urinary malonyldialdehyde, elevated relative kidney weight, glomerulosclerosis, and increased glomerular size, but no relevant interstitial fibrosis. Neither dose of Rlx affected these changes although the drug was active and targeted plasma levels were achieved. Of note, we found no activation of the renal TGF-ß pathway in this model. In the hearts of diabetic animals, no fibrotic alterations indicative of DC could be determined which precluded testing of the initial hypothesis. CONCLUSIONS: We investigated a model showing early DN without overt tubulointerstitial fibrosis and activation of the TGF-ß-Smad-2/3 pathway. In this model, Rlx proved ineffective; however, the same may not apply to other models and types of diabetes.


Subject(s)
Diabetes Mellitus, Type 1/drug therapy , Diabetic Nephropathies/drug therapy , Disease Models, Animal , Relaxin/therapeutic use , Animals , Diabetes Mellitus, Type 1/pathology , Diabetic Nephropathies/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
6.
Transfus Med Hemother ; 42(1): 29-37, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25960713

ABSTRACT

BACKGROUND: Hematopoietic stem and progenitor cell (HPC) motility is essential for HPC transplantation. The chemokine CXCL12 is key for HPC motility. Further regulators are of interest to improve HPC transplantation and regenerative medicine. Here the impact of the human chemokine CCL15 on HPC motility was investigated. METHODS: CCL15 plasma concentrations were determined during HPC mobilization in humans. Activity of CCL15 on HPCs was investigated in murine assays, including chemotaxis, adhesion, and CFU-A assays, and competitive repopulation assays. RESULTS: During HPC mobilization with granulocyte colony-stimulating factor, blood plasma contains increased concentrations (1.1 ± 0.1 ng/ml) of activated CCL15(27-92) versus 0.4 ± 0.1 ng/ml in controls (p = 0.02). CCL15(27-92) significantly enhanced CXCL12-induced transwell migration of Lin-/Sca1+ HPCs and strengthened shear stress-dependent adhesion to vascular cell adhesion molecule-1 (VCAM-1). CCL15(27-92) dose-dependently reduced the colony size in CFU-A assays performed with murine bone marrow and Lin-/Sca1+ HPCs. CCL15(27-92) did not show a direct impact on cell cycle status of HPCs. In murine repopulation assays, pretreatment of bone marrow with CCL15(27-92) significantly increased competitive repopulation. CONCLUSION: Our results point to a regulation of HPCs by CCL15 by modulating migratory and adhesive properties of HPCs with the potency to improve HPC short-term engraftment in stem cell transplantation.

7.
J Dermatol Sci ; 115(1): 13-20, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38849289

ABSTRACT

BACKGROUND: Lympho-epithelial Kazal-type-related inhibitor (LEKTI) is a serine protease inhibitor consisting of multiple domains. A loss of function mutation is described in Netherton patients that show severe symptoms of atopic lesions and itch. OBJECTIVES: LEKTI domain 6 (LD6) has shown strong serine protease-inhibitory action in in vitro assays and thus it was tested in vitro and in vivo for potential anti-inflammatory action in models of atopic skin disease. METHODS: Human skin equivalents were treated with LD6 and an inflammatory reaction was challenged by kallikrein-related endopeptidase 5 (KLK5). Furthermore, LD6 was tested on dorsal root ganglia cells stimulated with KLK5, SLIGRL and histamine by calcium imaging. The effect of topically administered LD6 (0.4-0.8%) in lipoderm was compared to a topical formulation of betamethasone-diproprionate (0.1%) in a therapeutic setting on atopic dermatitis-like lesions in NC/Nga mice sensitized to house dust mite antigen. Endpoints were clinical scoring of the mice as well as determination of scratching behaviour. RESULTS: KLK5 induced an upregulation of CXCL-8, CCL20 and IL-6 in skin equivalents. This upregulation was reduced by pre-incubation with LD6. KLK5 as well as histamine induced calcium influx in a population of neurons. LD6 significantly reduced the calcium response to both stimuli. When administered onto lesional skin of NC/Nga mice, both LD6 and betamethasone-dipropionate significantly reduced the inflammatory reaction. The effect on itch behaviour was less pronounced. CONCLUSION: Topical administration of LD6 might be a new therapeutic option for treatment of lesional atopic skin.


Subject(s)
Anti-Inflammatory Agents , Dermatitis, Atopic , Disease Models, Animal , Animals , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/immunology , Dermatitis, Atopic/pathology , Mice , Humans , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/administration & dosage , Skin/drug effects , Skin/pathology , Skin/immunology , Pruritus/drug therapy , Pruritus/immunology , Pruritus/pathology , Serine Peptidase Inhibitor Kazal-Type 5/metabolism , Serine Peptidase Inhibitor Kazal-Type 5/genetics , Serine Peptidase Inhibitor Kazal-Type 5/immunology , Kallikreins/metabolism , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Female , Interleukin-6/metabolism , Administration, Cutaneous
8.
Peptides ; 175: 171111, 2024 May.
Article in English | MEDLINE | ID: mdl-38036098

ABSTRACT

Endogenous peptide inhibitor for CXCR4 (EPI-X4) is a CXCR4 antagonist with potential for cancer therapy. It is a processed fragment of serum albumin from the hemofiltrate of dialysis patients. This study reports the efficacy of fifteen EPI-X4 derivatives in pancreatic cancer and lymphoma models. In vitro, the peptides were investigated for antiproliferation (cytotoxicity) by MTT assay. The mRNA expression for CXCR4 and CXCL12 was determined by RT-PCR, chip array and RNA sequencing. Chip array analysis yielded 634 genes associated with CXCR4/CXCL12 signaling. About 21% of these genes correlated with metastasis in the context of cell motility, proliferation, and survival. Expression levels of these genes were altered in pancreatic cancer (36%), lymphoma models (53%) and in patients' data (58%). EPI-X4 derivatives failed to inhibit cell proliferation due to low expression of CXCR4 in vitro, but inhibited tumor growth in the bioassays with significant efficacy. In the pancreatic cancer model, EPI-X4a, f and k inhibited mean tumor growth by > 50% and even caused complete remissions. In the lymphoma model, EPI-X4b, n and p inhibited mean tumor growth by > 70% and caused stable disease. Given the non-toxic and non-immunogenic properties of EPI-X4, these findings underscore its status as a promising therapy of pancreatic cancer and lymphoma and warrant further studies. SIMPLE SUMMARY: This study examined the value of chemokine receptor CXCR4 as an antineoplastic target for the endogenous peptide inhibitor of CXCR4 (EPI-X4), a 12-meric peptide derived from serum albumin. EPI-X4 inhibits CXCR4 interaction with its natural ligand, CXCL12 (SDF1). Therefore, malignancies (including pancreatic cancer and lymphoma) that depend on the CXCR4/CXCL12 pathway for progression can be targeted with EPI-X4. Of 634 genes that were linked to the CXCR4/CXCL12 pathway, 21% were associated with metastasis. In cultured human Suit2-007 pancreatic cancer cells, CXCR4 showed low to undetectable expression, which was why EPI-X4 did not inhibit pancreatic cancer cell proliferation. These findings were different in vivo, where CXCR4 was highly expressed and EPI-X4 inhibited tumor growth in rodents harboring pancreatic cancer or lymphoma. In the pancreatic cancer model, EPI-X4 derivatives a, f and k caused complete remissions, while in lymphomas EPI-X4 derivatives b, n and p caused stable disease.


Subject(s)
Lymphoma , Pancreatic Neoplasms , Humans , Cell Line, Tumor , Cell Proliferation , Lymphoma/drug therapy , Lymphoma/genetics , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Peptides/chemistry , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Serum Albumin/chemistry , Serum Albumin/metabolism , Signal Transduction
9.
Antimicrob Agents Chemother ; 57(10): 4751-60, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23856778

ABSTRACT

Naturally occurring substances with antimicrobial activity can serve as a starting point for the rational design of new drugs to treat infectious diseases. Here, we screened a library of peptides derived from human hemofiltrate for inhibitory effects on human cytomegalovirus (CMV) infection. We isolated a previously unknown derivative of the neutrophil-activating peptide 2, which we termed CYVIP, for CMV-inhibiting peptide. The peptide blocked infection with human and mouse CMV as well as with herpes simplex virus type 1 in different cell types. We found that CYVIP interferes with virus attachment to the cell surface, and structure-activity relationship studies revealed that positively charged lysine and arginine residues of CYVIP are essential for its inhibitory activity. The N-terminal 29 amino acids of the peptide were sufficient for inhibition, and substitution with an acidic residue further improved its activity. The target structure of CYVIP on the cell surface seems to be the sulfate residues of heparan sulfate proteoglycans, which are known to serve as herpesvirus attachment receptors. Our data suggest that O-sulfation of heparan sulfate is required for binding of CYVIP, and furthermore, that the initial interaction of CMV particles with cells takes place preferentially via 6-O-linked sulfate groups. These findings about CYVIP's mode of action lay the basis for further development of antivirals interfering with attachment of CMV to cells, a crucial step of the infection cycle.


Subject(s)
Cytomegalovirus/chemistry , Heparan Sulfate Proteoglycans/chemistry , Herpesvirus 1, Human/chemistry , Fluorometry , Humans , beta-Thromboglobulin/chemistry
10.
J Virol ; 86(2): 1244-9, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22090109

ABSTRACT

Semen is the major vector for HIV-1 transmission. We previously isolated C-proximal fragments of the prostatic acid phosphatase (PAP) from semen which formed amyloid fibrils that potently enhanced HIV infection. Here, we used the same methodology and identified another amyloidogenic peptide. Surprisingly, this peptide is derived from an N-proximal fragment of PAP (PAP85-120) and forms, similar to the C-proximal fragments, positively charged fibrillar structures that increase virion attachment to cells. Our results provide a first example for amyloid formation by fragments of distinct regions of the same precursor and further emphasize the possible importance of amyloidogenic peptides in HIV transmission.


Subject(s)
Amyloid/metabolism , HIV Infections/enzymology , HIV-1/physiology , Peptide Fragments/metabolism , Protein Tyrosine Phosphatases/chemistry , Protein Tyrosine Phosphatases/metabolism , Semen/enzymology , Acid Phosphatase , Amino Acid Motifs , Amino Acid Sequence , Amyloid/chemistry , Cell Line , HIV Infections/transmission , HIV Infections/virology , Humans , Male , Molecular Sequence Data , Peptide Fragments/chemistry , Protein Tyrosine Phosphatases/genetics , Semen/chemistry , Sequence Alignment , Virus Attachment
11.
FASEB J ; 26(12): 5141-51, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22972919

ABSTRACT

APETx3, a novel peptide isolated from the sea anemone Anthopleura elegantissima, is a naturally occurring mutant from APETx1, only differing by a Thr to Pro substitution at position 3. APETx1 is believed to be a selective modulator of human ether-á-go-go related gene (hERG) potassium channels with a K(d) of 34 nM. In this study, APETx1, 2, and 3 have been subjected to an electrophysiological screening on a wide range of 24 ion channels expressed in Xenopus laevis oocytes: 10 cloned voltage-gated sodium channels (Na(V) 1.2-Na(V)1.8, the insect channels DmNa(V)1, BgNa(V)1-1a, and the arachnid channel VdNa(V)1) and 14 cloned voltage-gated potassium channels (K(V)1.1-K(V)1.6, K(V)2.1, K(V)3.1, K(V)4.2, K(V)4.3, K(V)7.2, K(V)7.4, hERG, and the insect channel Shaker IR). Surprisingly, the Thr3Pro substitution results in a complete abolishment of APETx3 modulation on hERG channels and provides this toxin the ability to become a potent (EC(50) 276 nM) modulator of voltage-gated sodium channels (Na(V)s) because it slows down the inactivation of mammalian and insect Na(V) channels. Our study also shows that the homologous toxins APETx1 and APETx2 display promiscuous properties since they are also capable of recognizing Na(V) channels with IC(50) values of 31 nM and 114 nM, respectively, causing an inhibition of the sodium conductance without affecting the inactivation. Our results provide new insights in key residues that allow these sea anemone toxins to recognize distinct ion channels with similar potency but with different modulatory effects. Furthermore, we describe for the first time the target promiscuity of a family of sea anemone toxins thus far believed to be highly selective.


Subject(s)
Ion Channel Gating/drug effects , Point Mutation , Sea Anemones/metabolism , Toxins, Biological/pharmacology , Animals , Cnidarian Venoms/genetics , Cnidarian Venoms/metabolism , Cnidarian Venoms/pharmacology , Dose-Response Relationship, Drug , Electrophysiology , Female , Humans , Insecta/genetics , Insecta/metabolism , Ion Channel Gating/physiology , Oocytes/drug effects , Oocytes/metabolism , Oocytes/physiology , Potassium Channels/genetics , Potassium Channels/metabolism , Potassium Channels/physiology , Sea Anemones/genetics , Sodium Channels/genetics , Sodium Channels/metabolism , Sodium Channels/physiology , Toxins, Biological/genetics , Toxins, Biological/metabolism , Xenopus laevis
12.
Amyloid ; 30(4): 424-433, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37431668

ABSTRACT

BACKGROUND: Systemic AA amyloidosis is a world-wide occurring protein misfolding disease in humans and animals that arises from the formation of amyloid fibrils from serum amyloid A (SAA) protein and their deposition in multiple organs. OBJECTIVE: To identify new agents that prevent fibril formation from SAA protein and to determine their mode of action. MATERIALS AND METHODS: We used a cell model for the formation of amyloid deposits from SAA protein to screen a library of peptides and small proteins, which were purified from human hemofiltrate. To clarify the inhibitory mechanism the obtained inhibitors were characterised in cell-free fibril formation assays and other biochemical methods. RESULTS: We identified lysozyme as an inhibitor of SAA fibril formation. Lysozyme antagonised fibril formation both in the cell model as well as in cell-free fibril formation assays. The protein binds SAA with a dissociation constant of 16.5 ± 0.6 µM, while the binding site on SAA is formed by segments of positively charged amino acids. CONCLUSION: Our data imply that lysozyme acts in a chaperone-like fashion and prevents the aggregation of SAA protein through direct, physical interactions.


Subject(s)
Amyloidosis , Immunoglobulin Light-chain Amyloidosis , Animals , Humans , Serum Amyloid A Protein/metabolism , Muramidase , Amyloidosis/metabolism , Amyloid/metabolism
13.
Am J Physiol Cell Physiol ; 303(12): C1260-8, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23054060

ABSTRACT

In this study, the interaction of natriuretic peptides (NP) and bradykinin (BK) signaling pathways was identified by measuring membrane potential (V(m)) and intracellular Ca(2+) using the patch-clamp technique and flow cytometry in HEK-293 cells. BK and NP receptor mRNA was identified using RT-PCR. BK (100 nM) depolarized cells activating bradykinin receptor type 2 (B(2)R) and Ca(2+)-dependent Cl(-) channels inhibitable by 5-nitro-2-(3-phenylpropylamino)benzoic acid (NPPB; 10 µM). The BK-induced Ca(2+) signal was blocked by the B(2)R inhibitor HOE 140. [Des-Arg(9)]-bradykinin, an activator of B(1)R, had no effect on intracellular Ca(2+). NP [atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), C-type natriuretic peptide (CNP), and urodilatin] depolarized HEK-293 cells inhibiting K(+) channels. ANP, urodilatin, BNP [binding to natriuretic peptide receptor (NPR)-A] and 8-bromo-(8-Br)-cGMP inhibited the BK-induced depolarization while CNP (binding to NPR-Bi) failed to do so. The inhibitory effect on BK-triggered depolarization could be reversed by blocking PKG using the specific inhibitor KT 5823. BK-stimulated depolarization as well as Ca(2+) signaling was completely blocked by the phospholipase C (PLC) inhibitor U-73122 (10 nM). The inositol 1,4,5-trisphosphate receptor blocker 2-aminoethoxydiphenyl borate (2-APB; 50 µM) completely inhibited the BK-induced Ca(2+) signaling. UTP, another activator of the PLC-mediated Ca(2+) signaling pathway, was blocked by U-73122 as well but not by 8-Br-cGMP, indicating an intermediate regulatory step for NP via PKG in BK signaling such as regulators of G-protein signaling (RGS) proteins. When RGS proteins were inhibited by CCG-63802 in the presence of BK and 8-Br-cGMP, cells started to depolarize again. In conclusion, as natural antagonists of the B(2)R signaling pathway, NP may also positively interact in pathological conditions caused by BK.


Subject(s)
Bradykinin/pharmacology , Natriuretic Peptides/pharmacology , RGS Proteins/antagonists & inhibitors , Boron Compounds , Bradykinin/analogs & derivatives , Bradykinin B2 Receptor Antagonists , Carbazoles/pharmacology , Chloride Channels/antagonists & inhibitors , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Estrenes/pharmacology , Flow Cytometry , HEK293 Cells , Humans , Inositol 1,4,5-Trisphosphate Receptors/antagonists & inhibitors , Membrane Potentials/drug effects , Nitrobenzoates/pharmacology , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrrolidinones/pharmacology , RGS Proteins/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Thionucleotides/pharmacology , Type C Phospholipases/antagonists & inhibitors
14.
Biol Chem ; 392(10): 887-95, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21812756

ABSTRACT

Genetic modification of human embryonic stem cells (hESCs) using biophysical DNA transfection methods are hampered by the very low single cell survival rate and cloning efficiency of hESCs. Lentiviral gene transfer strategies are widely used to genetically modify hESCs but limited transduction efficiencies in the presence of feeder or stroma cells present problems, particularly if vesicular stomatitis virus glycoprotein (VSV-G) pseudotyped viral particles are applied. Here, we investigated whether the recently described semen derived enhancer of virus infection (SEVI) and alternative viral envelope proteins derived from either Gibbon ape leukaemia virus (GALV) or feline leukaemia virus (RD114) are applicable for transducing hESCs during co-culture with feeder or stroma cells. Our first set of experiments demonstrates that SEVI has no toxic effect on murine or hESCs and that exposure to SEVI does not interfere with the pluripotency-associated phenotype. Focusing on hESCs, we were able to further demonstrate that SEVI increases the transduction efficiencies of GALV and RD114 pseudotyped lentiviral vectors. More importantly, aiming at targeted differentiation of hESCs into functional somatic cell types, GALV pseudotyped lentiviral particles could efficiently and exclusively transduce hESCs grown in co-culture with OP9-GFP stroma cells (which were often used to induce differentiation into haematopoietic derivatives).


Subject(s)
Coculture Techniques , Embryonic Stem Cells/metabolism , Lentivirus/genetics , Leukemia Virus, Gibbon Ape/metabolism , Stromal Cells/metabolism , Transduction, Genetic/methods , Viral Envelope Proteins/metabolism , Animals , Cell Line , Coculture Techniques/methods , Embryonic Stem Cells/cytology , Genetic Vectors/genetics , Humans , Mice , Stromal Cells/cytology
15.
J Immunol ; 183(2): 1229-37, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19553544

ABSTRACT

The CC chemokine CCL14a is constitutively expressed in a large variety of tissues and its inactive proform CCL14a(1-74) circulates in high concentrations in plasma. CCL14a(1-74) is converted into CCL14a(9-74) by the proteases urokinase-type plasminogen activator and plasmin and is a highly active agonist for the chemokine receptors CCR1 and CCR5. In this study, a new CCL14a analog, CCL14a(12-74), was isolated from blood filtrate. To elucidate the functional role of the N terminus, a panel of N-terminally truncated CCL14a analogs were tested on the receptors CCR1 to CCR5 and on the human cytomegalovirus (HCMV)-encoded chemokine receptor US28. The rank order of binding affinity to these receptors and of the activation of CCR1 and CCR5-mediated intracellular Ca(2+) concentration mobilization is CCL14a(6-74)<(7-74)<(8-74)<<(9-74) = (10-74)>>(11-74)>>(12-74). The almost identical affinities of CCL14a(7-74), CCL14a(9-74), and CCL14a(10-74) for the US28 receptor and the inhibition of US28-mediated HIV infection of 293T cells by all of the N-terminally truncated CCL14a analogs support the promiscuous nature of the viral chemokine receptor US28. In high concentrations, CCL14a(12-74) did reveal antagonistic activity on intracellular Ca(2+) concentration mobilization in CCR1- and CCR5-transfected cells, which suggests that truncation of Tyr(11) might be of significance for an efficient inactivation of CCL14a. A putative inactivation pathway of CCL14a(9-74) to CCL14a(12-74) may involve the dipeptidase CD26/dipeptidyl peptidase IV (DPPIV), which generates CCL14a(11-74), and the metalloprotease aminopeptidase N (CD13), which displays the capacity to generate CCL14a(12-74) from CCL14a(11-74). Our results suggest that the activity of CCL14a might be regulated by stringent proteolytic activation and inactivation steps.


Subject(s)
Chemokines, CC/metabolism , Peptide Fragments/physiology , Peptide Hydrolases/metabolism , Receptors, Chemokine/metabolism , Calcium Signaling , Cell Line , Cytomegalovirus , Dipeptidyl Peptidase 4/metabolism , Fibrinolysin/metabolism , HIV Infections/prevention & control , Humans , Peptide Fragments/chemistry , Protein Binding , Receptors, CCR1/metabolism , Receptors, CCR5/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Viral Proteins/metabolism
16.
Front Oncol ; 11: 689063, 2021.
Article in English | MEDLINE | ID: mdl-34222016

ABSTRACT

Rearrangements in the Mixed Lineage Leukemia breakpoint cluster region (MLLbcr) are frequently involved in therapy-induced leukemia, a severe side effect of anti-cancer therapies. Previous work unraveled Endonuclease G as the critical nuclease causing initial breakage in the MLLbcr in response to different types of chemotherapeutic treatment. To identify peptides protecting against therapy-induced leukemia, we screened a hemofiltrate-derived peptide library by use of an enhanced green fluorescent protein (EGFP)-based chromosomal reporter of MLLbcr rearrangements. Chromatographic purification of one active fraction and subsequent mass spectrometry allowed to isolate a C-terminal 27-mer of fibrinogen α encompassing amino acids 603 to 629. The chemically synthesized peptide, termed Fα27, inhibited MLLbcr rearrangements in immortalized hematopoietic cells following treatment with the cytostatics etoposide or doxorubicin. We also provide evidence for protection of primary human hematopoietic stem and progenitor cells from therapy-induced MLLbcr breakage. Of note, fibrinogen has been described to activate toll-like receptor 4 (TLR4). Dissecting the Fα27 mode-of action revealed association of the peptide with TLR4 in an antagonistic fashion affecting downstream NFκB signaling and pro-inflammatory cytokine production. In conclusion, we identified a hemofiltrate-derived peptide inhibitor of the genome destabilizing events causing secondary leukemia in patients undergoing chemotherapy.

17.
Retrovirology ; 7: 55, 2010 Jun 23.
Article in English | MEDLINE | ID: mdl-20573198

ABSTRACT

BACKGROUND: HIV-1 is usually transmitted in the presence of semen. We have shown that semen boosts HIV-1 infection and contains fragments of prostatic acid phosphatase (PAP) forming amyloid aggregates termed SEVI (semen-derived enhancer of viral infection) that promote virion attachment to target cells. Despite its importance for the global spread of HIV-1, however, the effect of semen on virus infection is controversial. RESULTS: Here, we established methods allowing the meaningful analysis of semen by minimizing its cytotoxic effects and partly recapitulating the conditions encountered during sexual HIV-1 transmission. We show that semen rapidly and effectively enhances the infectivity of HIV-1, HIV-2, and SIV. This enhancement occurs independently of the viral genotype and coreceptor tropism as well as the virus producer and target cell type. Semen-mediated enhancement of HIV-1 infection was also observed under acidic pH conditions and in the presence of vaginal fluid. We further show that the potency of semen in boosting HIV-1 infection is donor dependent and correlates with the levels of SEVI. CONCLUSIONS: Our results show that semen strongly enhances the infectivity of HIV-1 and other primate lentiviruses and that SEVI contributes to this effect. Thus, SEVI may play an important role in the sexual transmission of HIV-1 and addition of SEVI inhibitors to microbicides may improve their efficacy.


Subject(s)
Amyloid/metabolism , HIV Infections/transmission , HIV-1/pathogenicity , Protein Tyrosine Phosphatases/metabolism , Semen/virology , Sexually Transmitted Diseases, Viral , Virus Attachment , Acid Phosphatase , Animals , Cell Line , HIV-2/pathogenicity , Humans , Hydrogen-Ion Concentration , Primates , Protein Binding , Simian Immunodeficiency Virus/pathogenicity
18.
J Gene Med ; 12(2): 137-46, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20052742

ABSTRACT

BACKGROUND: An improvement of retroviral infection has been postulated using a naturally occurring fragment of the abundant semen marker prostatic acidic phosphatase. This peptide, termed semen-derived enhancer of virus infection (SEVI), promotes HIV attachment to the target cells. METHODS: In the present study, we examined whether SEVI would also enhance the infectivity of other viruses with different envelope proteins. We focused on retroviruses pseudotyped with envelopes that are commonly used for the genetic modification of cells, in particular, T cells and hematopoietic progenitor cells. Because the effect of SEVI is considered to be a result of its cationic properties, we compared SEVI with other cationic agents such as protamine sulfate and Polybrene. RESULTS: We found that SEVI increases the efficiency of gene transfer for lentiviral and gammaretroviral vector constructs pseudotyped with VSV-G, GALV, RD114 or foamy viral envelopes on hematopoietic and nonhematopoietic cell lines. On T cells, the transduction efficiency of GALV and RD114 pseudotyped vectors was significantly increased by SEVI. A significant increase of the gene transfer rate was also detected for foamy virally pseudotyped lentivirus on murine hematopoietic progenitor cells. No toxic effect of SEVI treatment was detected on any cell type tested, including human and murine hematopoietic stem/progenitor cells. When directly comparing the effect of SEVI with Polybrene or protamine sulfate, we show that the semen-derived protein is more efficient in increasing the gene transfer rate. CONCLUSIONS: SEVI is a promising agent for promoting and improving gene transfer and may also be useful for clinical gene therapy studies.


Subject(s)
Gene Transfer Techniques , Peptide Fragments/pharmacology , Protein Tyrosine Phosphatases/pharmacology , Retroviridae/genetics , Acid Phosphatase , Animals , Antigens, CD34/metabolism , Cations , Cell Adhesion/drug effects , Cell Death/drug effects , Flow Cytometry , HeLa Cells , Hematopoietic System/cytology , Hematopoietic System/drug effects , Humans , Male , Mice , NIH 3T3 Cells , Peptide Fragments/chemistry , Protein Tyrosine Phosphatases/chemistry , Retroviridae/drug effects , Transduction, Genetic , Viral Envelope Proteins/metabolism
19.
Anal Biochem ; 401(1): 53-60, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20188690

ABSTRACT

A fragment of the human beta-chain of hemoglobin (HEM), hHEMbeta111-146, was shown to have broad antimicrobial properties. The 3.9-kDa peptide was postulated to occur in high concentrations in placenta tissue. We established a reliable method to quantify hHEMbeta111-146 in placenta tissue. Our methodology consists of a tissue extraction step (step 1), a chromatographic enrichment step (step 2), and a final quantification step (step 3) by enzyme-linked immunosorbent assay (ELISA). The specificity of the ELISA reaction was confirmed by parallel analysis of the samples via Western blot (step 4). The ELISA measured the absorbance of a tetramethylbenzidine substrate at 450nm. It showed no cross-reactivity with the corresponding gamma- and alpha-HEM regions and low cross-reactivity with the beta-HEM region and full-length HEM. The sample preparation procedure enabled a prepurification of hHEMbeta111-146, completely eliminating cross-reactive proteins and HEM peptides. The linear range of detection in step 3 was 20-200ng/well (200-2000microg/L) with a limit of quantification of 23ng/well (230microg/L) and a limit of detection of 7ng/well (70microg/L). The assay was characterized by good linearity (r(2)>0.99), intraday precision (coefficient of variation [CV]=2.2-8.3%), interday precision (CV=1.8-9.1%), and accuracy (76-109%). The mean recovery of the ELISA was determined to be 97%, and the overall recovery during steps 1-3 was found to be 40.3+/-2.5%. We measured concentrations from 0.28 to 0.74mg/g placenta tissue of the hHEMbeta111-146 in different placenta samples with an average concentration of 0.57mg/g. This abundant concentration supports an important physiological role of hHEMbeta111-146 in the placenta infective barrier.


Subject(s)
Antimicrobial Cationic Peptides/analysis , Enzyme-Linked Immunosorbent Assay/methods , Hemoglobins/metabolism , Placenta/metabolism , Amino Acid Sequence , Antimicrobial Cationic Peptides/isolation & purification , Blotting, Western , Female , Humans , Mass Spectrometry , Molecular Sequence Data , Pregnancy
20.
J Immunol ; 181(2): 1120-7, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18606664

ABSTRACT

Chemokines mediate the recruitment of leukocytes to the sites of inflammation. N-terminal truncation of chemokines by the protease dipeptidyl peptidase IV (DPPIV) potentially restricts their activity during inflammatory processes such as allergic reactions, but direct evidence in vivo is very rare. After demonstrating that N-terminal truncation of the chemokine CCL11/eotaxin by DPPIV results in a loss of CCR3-mediated intracellular calcium mobilization and CCR3 internalization in human eosinophils, we focused on the in vivo role of CCL11 and provide direct evidence for specific kinetic and rate-determining effects by DPPIV-like enzymatic activity on CCL11-mediated responses of eosinophils. Namely, it is demonstrated that i.v. administration of CCL11 in wild-type F344 rats leads to mobilization of eosinophils into the blood, peaking at 30 min. This mobilization is significantly increased in DPPIV-deficient F344 rats. Intradermal administration of CCL11 is followed by a dose-dependent recruitment of eosinophils into the skin and is significantly more effective in DPPIV-deficient F344 mutants as well as after pharmacological inhibition of DPPIV. Interestingly, CCL11 application leads to an up-regulation of DPPIV, which is not associated with negative feedback inhibition via DPPIV-cleaved CCL11((3-74)). These findings demonstrate regulatory effects of DPPIV for the recruitment of eosinophils. Furthermore, they illustrate that inhibitors of DPPIV have the potential to interfere with chemokine-mediated effects in vivo including but not limited to allergy.


Subject(s)
Chemokine CCL11/immunology , Chemotaxis, Leukocyte , Dipeptidyl Peptidase 4/metabolism , Eosinophils/immunology , Animals , Chemokine CCL11/metabolism , Dipeptidyl Peptidase 4/immunology , Dipeptidyl-Peptidase IV Inhibitors , Down-Regulation , Enzyme Inhibitors/pharmacology , Eosinophils/metabolism , Humans , Isoleucine/analogs & derivatives , Isoleucine/pharmacology , Rats , Rats, Inbred F344 , Rats, Mutant Strains , Receptors, CCR3/metabolism , Skin/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Thiazoles/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL