Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Biochem Pharmacol ; 188: 114565, 2021 06.
Article in English | MEDLINE | ID: mdl-33872569

ABSTRACT

The human CC chemokine receptor 8 (CCR8) is a promising drug target for cancer immunotherapy and autoimmune disease. Besides human and viral chemokines, previous studies revealed diverse classes of CCR8-targeting small molecules. We characterized a selection of these CCR8 ligands (hCCL1, vCCL1, ZK756326, AZ6; CCR8 agonists and a naphthalene-sulfonamide-based CCR8 antagonist), in in vitro cell-based assays (hCCL1AF647 binding, calcium mobilization, cellular impedance, cell migration, ß-arrestin 1/2 recruitment), and used pharmacological tools to determine G protein-dependent and -independent signaling pathways elicited by these ligands. Our data reveal differences in CCR8-mediated signaling induced by chemokines versus small molecules, which was most pronounced in cell migration studies. Human CCL1 most efficiently induced cell migration whereby Gßγ signaling was indispensable. In contrast, Gßγ signaling did not contribute to cell migration induced by other CCR8 ligands (vCCL1, ZK756326, AZ6). Although all tested CCR8 agonists were full agonists for calcium mobilization, a significant contribution for Gßγ signaling herein was only apparent for human and viral CCL1. Despite both Gαi- and Gαq-signaling regulate intracellular Ca2+-release, cellular impedance experiments showed that CCR8 agonists predominantly induce Gαi-dependent signaling. Finally, small molecule agonists displayed higher efficacy in ß-arrestin 1 recruitment, which occurred independently of Gαi signaling. Also in this latter assay, only hCCL1-induced activity was dependent on Gßγ-signaling. Our study provides insight into CCR8 signaling and function and demonstrates differential CCR8 activation by different classes of ligands. This reflects the ability of CCR8 small molecules to evoke different subsets of the receptor's signaling repertoire, which categorizes them as biased agonists.


Subject(s)
Drug Delivery Systems/methods , Receptors, CCR8/agonists , Receptors, CCR8/antagonists & inhibitors , Signal Transduction/physiology , Chemokine CCL1/administration & dosage , Chemokine CCL1/metabolism , Chemokine CCL8/administration & dosage , Chemokine CCL8/metabolism , Chemokines, CC/administration & dosage , Chemokines, CC/metabolism , Dose-Response Relationship, Drug , Humans , Jurkat Cells , Ligands , Receptors, CCR8/metabolism , Signal Transduction/drug effects
2.
Vaccine ; 38(11): 2626-2635, 2020 03 04.
Article in English | MEDLINE | ID: mdl-32057572

ABSTRACT

An effective prophylactic vaccine targeting HIV must induce a robust humoral response and must direct the bulk of this response to the mucosa-the primary site of HIV transmission. The chemokine, CCL28, is secreted by epithelial cells at mucosal surfaces and recruits' cells expressing its receptor CCR10. CCR10 is predominantly expressed by IgA + ASCs. We hypothesized that co-immunization with plasmid DNA encoding consensus envelope antigens with plasmid-encoded CCL28 would enhance anti-HIV IgA responses at mucosal surfaces. Indeed, animals receiving pCCL28 and pEnvA/C had significantly increased HIV-specific IgA in fecal extract. Surprisingly, CCL28 co-immunization induced a significant increase in anti-HIV IgG in the serum in mice compared to those receiving pEnvA/C alone. These robust antibody responses were not associated with changes in the frequency of germinal center B cells but depended upon the expression of CCR10, as these responses we abolished in CCR10-deficient animals. Finally, immunization with CCL28 led to increased frequencies in HIV-specific CCR10 + and CCR10 + IgA + B cells in the small intestine and Peyer's patches of vaccinated animals as compared to those receiving pEnvA/C alone. These data indicate that CCL28 administration can enhance antigen-specific humoral responses systemically and at mucosal surfaces.


Subject(s)
AIDS Vaccines/administration & dosage , Adjuvants, Immunologic/administration & dosage , Chemokines, CC/administration & dosage , Receptors, CCR10/genetics , Vaccines, DNA/administration & dosage , Animals , HIV Antibodies/immunology , HIV-1/immunology , Immunity, Mucosal , Immunoglobulin A/immunology , Immunoglobulin G/blood , Mice , Mucous Membrane/immunology , env Gene Products, Human Immunodeficiency Virus/immunology
3.
Aging (Albany NY) ; 11(18): 7402-7415, 2019 09 26.
Article in English | MEDLINE | ID: mdl-31557129

ABSTRACT

BACKGROUND: Chemokines play a key role in post-traumatic inflammation and secondary injury after spinal cord injury (SCI). CCL28, the chemokine CC-chemokine ligand 28, is involved in the epithelial and mucosal immunity. However, whether CCL28 participates in the physiopathologic processes after SCI remains unclear. RESULTS: CCL28 is upregulated in the spinal cord after SCI. In addition, neutralizing antibodies against IL-1ß or TNF-α, or treatment of ML120B, a selective inhibitor of IKK-ß, remarkably decrease CCL28 upregulation, suggesting that CCL28 upregulation relies on NF-κB pathway activated by IL-1ß and TNF-α after SCI. Moreover, CD4+CD25+FOXP3+ regulatory T (Treg) cells that express CCR10, a receptor of CCL28, are enriched in the spinal cord after SCI. We further demonstrate that the spinal cord recruits Treg cells through CCL28-CCR10 axis, which in turn function to suppress immune response and promote locomotor recovery after SCI. In contrast, neutralizing CCL28 or CCR10 reduces Treg cell recruitment and delays locomotor recovery. METHODS: The neutralizing antibodies and recombinant CCL28 were injected intraspinally into the mice prior to SCI, which was established via hemitransection. RT-qPCR analysis was performed to determine transcript level, and Western blot analysis and ELISA assay were used to detect protein expression. Immune cells were analyzed by flow cytometry and visualized by immunofluorescence. The chemotaxis was assessed by in vitro transwell migration assay. The mouse locomotor activity was assessed via the Basso Mouse Scale (BMS) system. CONCLUSIONS: These results indicate that NF-κB pathway-regulated CCL28 production plays a protective role after SCI through recruiting CCR10-expressing and immunosuppressive Treg cells, and suggest that interfering CCL28-CCR10 axis might be of potential clinical benefit in improving SCI recovery.


Subject(s)
Chemokines, CC/administration & dosage , Chemokines, CC/metabolism , Locomotion/physiology , T-Lymphocytes, Regulatory/physiology , Animals , Antibodies, Neutralizing , Gene Expression Regulation/drug effects , Interleukin-1beta/pharmacology , Locomotion/drug effects , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , Receptors, CCR10/genetics , Receptors, CCR10/metabolism , Recombinant Proteins , Spinal Cord Injuries , T-Lymphocytes, Regulatory/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation
4.
Physiol Behav ; 208: 112581, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31220516

ABSTRACT

FAM19A2/TAFA-2, a member of the chemokine CC family, shares 31% sequence identity with MIP-1α, which is known to elevate body temperature and reduce food intake. A single administration of 250 pM of FAM19A2/TAFA-2 to the third ventricle of mice just before the initiation of dark period increased food intake and meal number significantly, but reduced meal size during the dark period. The respiratory exchange rate and energy expenditure were increased significantly during the dark period, while the ambulatory count and vertical activity were not affected. These data suggest that FAM19A2/TAFA-2 participates in the regulation of food intake and metabolic activities.


Subject(s)
Activation, Metabolic/physiology , Chemokines, CC/physiology , Eating/physiology , Activation, Metabolic/drug effects , Animals , Body Temperature/drug effects , Body Temperature/physiology , Chemokines, CC/administration & dosage , Chemokines, CC/pharmacology , Circadian Rhythm , Eating/drug effects , Infusions, Intraventricular , Male , Mice
5.
Int J Pharm ; 564: 207-213, 2019 Jun 10.
Article in English | MEDLINE | ID: mdl-30999049

ABSTRACT

The past fifteen years have witnessed a resurgence of interest in vaginal ring technologies for drug delivery applications, mostly driven by the impetus for development of vaginally-administered antiretroviral microbicides to help reduce the high acquisition rates for human immunodeficiency virus (HIV) among Sub-Saharan African women. Currently, the lead candidate microbicide is a 28-day silicone elastomer vaginal ring releasing dapivirine (Ring-004), an experimental non-nucleoside reverse transcriptase inhibitor. The ring was tested in two pivotal Phase III clinical studies in 2016 and is currently undergoing review by the European Medicines Agency. Recently, we described a new type of silicone elastomer vaginal ring offering sustained release of the protein molecule 5P12-RANTES, a potent experimental chemokine analogue that potently blocks the HIV CCR5 coreceptor. Building on our previous work, here we report the preclinical development of a new combination vaginal ring that offers sustained release of both 5P12-RANTES and dapivirine, in which the 5P12-RANTES is incorporated into an exposed core within the ring body and the dapivirine in the sheath. In this way, in vitro release of dapivirine matches closely that for Ring-004. Also, we report the pharmacokinetic testing of this combination ring formulation in sheep, where vaginal concentrations of both drugs are maintained over 28 days at levels potentially useful for preventing HIV infection in women.


Subject(s)
Anti-HIV Agents/administration & dosage , Chemokines, CC/administration & dosage , Contraceptive Devices, Female , HIV Infections/prevention & control , Pyrimidines/administration & dosage , Animals , Anti-HIV Agents/pharmacokinetics , Chemokines, CC/pharmacokinetics , Delayed-Action Preparations/administration & dosage , Drug Combinations , Female , Pyrimidines/pharmacokinetics , Sheep , Vagina/metabolism
6.
J Control Release ; 301: 1-12, 2019 05 10.
Article in English | MEDLINE | ID: mdl-30876951

ABSTRACT

With almost 2 million new HIV infections worldwide each year, the prevention of HIV infection is critical for stopping the pandemic. The only approved form of pre-exposure prophylaxis is a costly daily pill, and it is recognized that several options will be needed to provide protection to the various affected communities around the world. In particular, many at-risk people would benefit from a prevention method that is simple to use and does not require medical intervention or a strict daily regimen. We show that silk fibroin protein can be formulated into insertable discs that encapsulate either an antibody (IgG) or the potent HIV inhibitor 5P12-RANTES. Several formulations were studied, including silk layering, water vapor annealing and methanol treatment to stabilize the protein cargo and impact the release kinetics over weeks. In the case of IgG, high concentrations were released over a short time using methanol treatment, with more sustained results with the use of water vapor annealing and layering during device fabrication. For 5P12-RANTES, sustained release was obtained for 31 days using water vapor annealing. Further, we show that the released inhibitor 5P12-RANTES was functional both in vitro and in ex vivo colorectal tissue. This work shows that silk fibroin discs can be developed into formidable tools to prevent HIV infection.


Subject(s)
Chemokines, CC/administration & dosage , Fibroins/administration & dosage , HIV Infections/prevention & control , Immunoglobulin G/administration & dosage , Cell Line , Chemokines, CC/chemistry , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/chemistry , Drug Liberation , Fibroins/chemistry , Humans , Immunoglobulin G/chemistry , Pre-Exposure Prophylaxis
7.
J Control Release ; 298: 1-11, 2019 03 28.
Article in English | MEDLINE | ID: mdl-30731150

ABSTRACT

Antiretroviral-releasing vaginal rings are at the forefront of ongoing efforts to develop microbicide-based strategies for prevention of heterosexual transmission of the human immunodeficiency virus (HIV). However, traditional ring designs are generally only useful for vaginal administration of relatively potent, lipophilic, and small molecular weight drug molecules that have sufficient permeability in the non-biodegradable silicone elastomer or thermoplastic polymers. Here, we report a novel, easy-to-manufacture 'exposed-core' vaginal ring that provides sustained release of the protein microbicide candidate 5P12-RANTES, an experimental chemokine analogue that potently blocks the HIV CCR5 coreceptor. In vitro release, mechanical, and stability testing demonstrated the utility and practicality of this novel ring design. In a sheep pharmacokinetic model, a ring containing two »-length excipient-modified silicone elastomer cores - each containing lyophilised 5P12-RANTES and exposed to the external environment by two large windows - provided sustained concentrations of 5P12-RANTES in vaginal fluid and vaginal tissue between 10 and 10,000 ng/g over 28days, at least 50 and up to 50,000 times the reported in vitro IC50 value.


Subject(s)
CCR5 Receptor Antagonists/administration & dosage , Chemokines, CC/administration & dosage , Contraceptive Devices, Female , Drug Delivery Systems , Administration, Intravaginal , Animals , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/pharmacokinetics , CCR5 Receptor Antagonists/pharmacokinetics , Chemokines, CC/pharmacokinetics , Delayed-Action Preparations , Female , HIV Infections/prevention & control , Humans , Inhibitory Concentration 50 , Sheep
8.
Int J Mol Sci ; 20(1)2018 Dec 23.
Article in English | MEDLINE | ID: mdl-30583576

ABSTRACT

Thymus-expressed chemokine (CCL25) is a potent cell attractant for mesenchymal stromal cells, and therefore it is a candidate for in situ cartilage repair approaches focusing on the recruitment of endogenous repair cells. However, the influence of CCL25 on cartilage is unknown. Accordingly, in this study, we investigated the effect of CCL25 on tissue-engineered healthy and osteoarthritic cartilage. Porcine chondrocytes were cultured in a three-dimensional (3D) micromass model that has been proven to mimic key-aspects of human cartilage and osteoarthritic alterations upon stimulation with tumor necrosis factor-α (TNF-α). Micromass cultures were stimulated with CCL25 (0, 0.05, 0.5, 5, 50, 500 nmol/L) alone or in combination with 0.6 nmol/L TNF-α for seven days. Effects were evaluated by life/dead staining, safranin O staining, histomorphometrical analysis of glycosaminoglycans (GAGs), collagen type II (COL2A1) real-time RT-PCR and Porcine Genome Array analysis. 500 nmol/L CCL25 led to a significant reduction of GAGs and COL2A1 expression and induced the expression of matrix metallopeptidases (MMP) 1, MMP3, early growth response protein 1 (EGR1), and superoxide dismutase 2 (SOD2). In concentrations lower than 500 nmol/L, CCL25 seems to be a candidate for in situ cartilage repair therapy approaches.


Subject(s)
Cartilage, Articular/drug effects , Chemokines, CC/administration & dosage , Mesenchymal Stem Cells/drug effects , Osteoarthritis/drug therapy , Tissue Engineering , Animals , Cartilage, Articular/metabolism , Chondrocytes/cytology , Chondrocytes/drug effects , Dose-Response Relationship, Drug , Female , Femur/cytology , Femur/drug effects , Humans , Mesenchymal Stem Cells/cytology , Osteoarthritis/metabolism , Primary Cell Culture , Recombinant Proteins/administration & dosage , Swine , Tumor Necrosis Factor-alpha/administration & dosage
9.
J Control Release ; 233: 208-19, 2016 07 10.
Article in English | MEDLINE | ID: mdl-27178810

ABSTRACT

Influenza infection typically initiates at respiratory mucosal surfaces. Induction of immune responses at the sites where pathogens initiate replication is crucial for the prevention of infection. We studied the adjuvanticity of GPI-anchored CCL28 co-incorporated with influenza HA-antigens in chimeric virus-like particles (cVLPs), in boosting strong protective immune responses through an intranasal (i.n.) route in mice. We compared the immune responses to that from influenza VLPs without CCL28, or physically mixed with soluble CCL28 at systemic and various mucosal compartments. The cVLPs containing GPI-CCL28 showed in-vitro chemotactic activity towards spleen and lung cells expressing CCR3/CCR10 chemokine receptors. The cVLPs induced antigen specific endpoint titers and avidity indices of IgG in sera and IgA in tracheal, lung, and intestinal secretions, significantly higher (4-6 fold) than other formulations. Significantly higher (3-5 fold) hemagglutination inhibition titers and high serum neutralization against H3N2 viruses were also detected with CCL28-containing VLPs compared to other groups. The CCL28-containing VLPs showed complete and 80% protection, when vaccinated animals were challenged with A/Aichi/2/1968/H3N2 (homologous) and A/Philippines/2/1982/H3N2 (heterologous) viruses, respectively. Thus, GPI-anchored CCL28 in influenza VLPs act as a strong immunostimulator at both systemic and mucosal sites, boosting significant cross-protection in animals against heterologous viruses across a large distance.


Subject(s)
Antigens, Viral/administration & dosage , Chemokines, CC/administration & dosage , GPI-Linked Proteins/administration & dosage , Hemagglutinins, Viral/administration & dosage , Influenza A Virus, H3N2 Subtype/immunology , Vaccines, Virus-Like Particle/administration & dosage , Viral Matrix Proteins/administration & dosage , Administration, Intranasal , Animals , Antibodies, Viral/immunology , Cell Line , Dogs , Female , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Madin Darby Canine Kidney Cells , Mice, Inbred BALB C , Orthomyxoviridae Infections/prevention & control , Spodoptera
10.
J Biol Chem ; 290(7): 4528-36, 2015 Feb 13.
Article in English | MEDLINE | ID: mdl-25556652

ABSTRACT

CCL28 is a human chemokine constitutively expressed by epithelial cells in diverse mucosal tissues and is known to attract a variety of immune cell types including T-cell subsets and eosinophils. Elevated levels of CCL28 have been found in the airways of individuals with asthma, and previous studies have indicated that CCL28 plays a vital role in the acute development of post-viral asthma. Our study builds on this, demonstrating that CCL28 is also important in the chronic post-viral asthma phenotype. In the absence of a viral infection, we also demonstrate that CCL28 is both necessary and sufficient for induction of asthma pathology. Additionally, we present the first effort aimed at elucidating the structural features of CCL28. Chemokines are defined by a conserved tertiary structure composed of a three-stranded ß-sheet and a C-terminal α-helix constrained by two disulfide bonds. In addition to the four disulfide bond-forming cysteine residues that define the traditional chemokine fold, CCL28 possesses two additional cysteine residues that form a third disulfide bond. If all disulfide bonds are disrupted, recombinant human CCL28 is no longer able to drive mouse CD4+ T-cell chemotaxis or in vivo airway hyper-reactivity, indicating that the conserved chemokine fold is necessary for its biologic activity. Due to the intimate relationship between CCL28 and asthma pathology, it is clear that CCL28 presents a novel target for the development of alternative asthma therapeutics.


Subject(s)
Asthma/pathology , CD4-Positive T-Lymphocytes/pathology , Chemokines, CC/chemistry , Chemokines, CC/metabolism , Epithelial Cells/pathology , Respirovirus Infections/pathology , Amino Acid Sequence , Animals , Asthma/immunology , Asthma/metabolism , Asthma/virology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Chemokines, CC/administration & dosage , Chemotaxis , Chronic Disease , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/virology , Humans , Magnetic Resonance Spectroscopy , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Protein Conformation , Respirovirus Infections/immunology , Respirovirus Infections/metabolism , Respirovirus Infections/virology , Sendai virus/pathogenicity , Sequence Homology, Amino Acid , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Structure-Activity Relationship , T-Lymphocyte Subsets
11.
Arch Dermatol Res ; 307(3): 275-80, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25384437

ABSTRACT

Inflammation in atopic dermatitis is mediated in part by the chemokine CCL1 and its receptor, CCR8. Recombinant Molluscum contagiosum viral protein (rMC148p), a cc-chemokine homolog, inhibits CCL1-induced chemotaxis of cells expressing CCR8. rMC148p was prepared using the baculovirus/Sf9 insect cell expression system. The recombinant MC148 fusion protein (rMC148fp), rMC148-TAT-6xHis, was similarly prepared by adding base sequences onto the PCR primers to fuse TAT and 6xHis to rMC148p at the carboxyl terminus. rMC148fp retains the capacity of rMC148p to inhibit CCL1-induced chemotaxis. Furthermore, unlike rMC148p, topically applied rMC148fp penetrates stratum corneum of human neonatal foreskins and concentrates along the basal and lower spinous cell layers of the epidermis. rMC148fp may be a safe and effective agent in the treatment of atopic dermatitis and other CCR8-mediated disorders.


Subject(s)
Chemokine CCL1/metabolism , Chemokines, CC/administration & dosage , Dermatitis, Atopic/therapy , Epidermis/drug effects , Immunotherapy , Receptors, CCR8/metabolism , Recombinant Fusion Proteins/administration & dosage , Administration, Topical , Animals , Baculoviridae/genetics , Chemotaxis/drug effects , Clinical Protocols , Cloning, Molecular , Dermatitis, Atopic/immunology , Epidermis/immunology , Foreskin/cytology , Humans , Infant, Newborn , Male , Molluscum Contagiosum/genetics , Molluscum Contagiosum/immunology , Recombinant Fusion Proteins/genetics , Sf9 Cells , Spodoptera
12.
Blood ; 124(2): 296-304, 2014 Jul 10.
Article in English | MEDLINE | ID: mdl-24876562

ABSTRACT

Development of T cells in the thymus requires continuous importation of T-lineage progenitors from the bone marrow via the circulation. Following bone marrow transplant, recovery of a normal peripheral T-cell pool depends on production of naïve T cells in the thymus; however, delivery of progenitors to the thymus limits T-lineage reconstitution. Here, we examine homing of intravenously delivered progenitors to the thymus following irradiation and bone marrow reconstitution. Surprisingly, following host conditioning by irradiation, we find that homing of lymphoid-primed multipotent progenitors and common lymphoid progenitors to the thymus decreases more than 10-fold relative to unirradiated mice. The reduction in thymic homing in irradiated mice is accompanied by a significant reduction in CCL25, an important chemokine ligand for thymic homing. We show that pretreatment of bone marrow progenitors with CCL25 and CCL21 corrects the defect in thymic homing after irradiation and promotes thymic reconstitution. These data suggest new therapeutic approaches to promote T-cell regeneration.


Subject(s)
Bone Marrow Transplantation , Cell Movement/drug effects , Chemokine CCL21/administration & dosage , Chemokines, CC/administration & dosage , Lymphoid Progenitor Cells/drug effects , T-Lymphocytes/drug effects , Transplantation Conditioning , Animals , Cell Lineage/drug effects , Cell Lineage/immunology , Cells, Cultured , Female , Lymphoid Progenitor Cells/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , T-Lymphocytes/physiology , Thymus Gland/cytology , Thymus Gland/drug effects , Thymus Gland/radiation effects
13.
Mol Pharm ; 10(10): 3564-73, 2013 Oct 07.
Article in English | MEDLINE | ID: mdl-23859720

ABSTRACT

5P12-RANTES is a recently developed chemokine analogue that has shown high level protection from SHIV infection in macaques. However, the feasibility of using 5P12-RANTES as a long-term HIV prevention agent has not been explored partially due to the lack of available delivery devices that can easily be modified for long-term release profiles. Glycosaminoglycans (GAGs) have been known for their affinity for various cytokines and chemokines, including native RANTES, or CCL5. In this work, we investigated used of GAGs in generating a chemokine drug delivery device. Initial studies used surface plasmon resonance analysis to characterize and compare the affinities of different GAGs to 5P12-RANTES. These different GAGs were then incorporated into drug delivery polymeric hydrogels to engineer sustained release of the chemokines. In vitro release studies of 5P12-RANTES from the resulting polymers were performed, and we found that 5P12-RANTES release from these polymers can be controlled by the amount and type of GAG incorporated. Polymer disks containing GAGs with stronger affinity to 5P12-RANTES resulted in more sustained and longer term release than did polymer disks containing GAGs with weaker 5P12-RANTES affinity. Similar trends were observed by varying the amount of GAGs incorporated into the delivery system. 5P12-RANTES released from these polymers demonstrated good levels of CCR5 blocking, retaining activity even after 30 days of incubation.


Subject(s)
Chemokines, CC/chemistry , Chemokines/chemistry , Drug Carriers/chemistry , Glycosaminoglycans/chemistry , HIV Infections/prevention & control , Polymers/chemistry , Chemokines, CC/administration & dosage , Humans , Surface Plasmon Resonance
14.
J Immunol ; 179(3): 1996-2004, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17641067

ABSTRACT

Expression of chemokine receptors by tumors, specifically CCR4 on cutaneous T cell lymphomas, is often associated with a poor disease outcome. To test the hypothesis that chemokine receptor-expressing tumors can be successfully controlled by delivering toxins through their chemokine receptors, we have generated fusion proteins designated chemotoxins: chemokines fused with toxic moieties that are nontoxic unless delivered into the cell cytosol. We demonstrate that chemokines fused with human RNase eosinophil-derived neurotoxin or with a truncated fragment of Pseudomonas exotoxin 38 are able to specifically kill tumors in vitro upon internalization through their respective chemokine receptors. Moreover, treatment with the thymus and activation-regulated chemokine (CCL17)-expressing chemotoxin efficiently eradicated CCR4-expressing cutaneous T cell lymphoma/leukemia established in NOD-SCID mice. Taken together, this work represents a novel concept that may allow control of growth and dissemination of tumors that use chemokine receptors to metastasize and circumvent immunosurveillance.


Subject(s)
Antineoplastic Agents/toxicity , Cytotoxicity, Immunologic , Immunotoxins/toxicity , Leukemia-Lymphoma, Adult T-Cell/immunology , Leukemia-Lymphoma, Adult T-Cell/therapy , Receptors, Chemokine/metabolism , ADP Ribose Transferases/genetics , ADP Ribose Transferases/toxicity , Animals , Antineoplastic Agents/metabolism , Bacterial Toxins/genetics , Bacterial Toxins/toxicity , Cell Death/genetics , Cell Death/immunology , Cell Line , Cell Line, Tumor , Chemokine CCL17 , Chemokines, CC/administration & dosage , Chemokines, CC/genetics , Chemokines, CC/toxicity , Cytotoxicity, Immunologic/genetics , Eosinophil-Derived Neurotoxin/genetics , Eosinophil-Derived Neurotoxin/toxicity , Exotoxins/genetics , Exotoxins/toxicity , Female , Humans , Immunotoxins/genetics , Leukemia-Lymphoma, Adult T-Cell/metabolism , Leukemia-Lymphoma, Adult T-Cell/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Recurrence, Local , Receptors, CCR4 , Receptors, Chemokine/biosynthesis , Viral Proteins/genetics , Viral Proteins/toxicity , Virulence Factors/genetics , Virulence Factors/toxicity , Pseudomonas aeruginosa Exotoxin A
15.
Cancer Gene Ther ; 14(6): 523-32, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17384577

ABSTRACT

Coexpression of tumor antigens together with immunomodulatory molecules is a strategy in DNA vaccination aiming at an amplification of the antitumor immune response. Epstein-Barr virus-induced-molecule-1-ligand-chemokine (ELC/CCL19) is a CC chemokine that binds to the chemokine receptor CCR7. CCR7 is expressed on mature dendritic cells (DC) and distinct T- and B-cell subpopulations. CCL19 (ELC) is mainly expressed in secondary lymphoid organs and plays a central role in regulating the encounters between DC and T cells. We asked whether CCL19 is able to augment immunogenicity of a DNA vaccine in a C57BL/6 mouse model with syngeneic MCA205 (beta-gal) tumor cells. Mice were vaccinated twice intramuscularly on days 1 and 15 and tumor challenge was performed subcutaneously on day 25. Coadministration of plasmid DNA (pDNA) (beta-gal) plus pDNA (CCL19) was compared with pDNA (beta-gal), pDNA (CCL19), mock vector and phosphate-buffered saline (PBS) alone. Coexpression of CCL19 resulted in enhancement of a Th1-polarized immune response with substantial improvement of the protective effect of the DNA vaccine. Immunohistochemical staining revealed an increased CD8+ T-cell infiltration in the tumor tissue of mice that had been immunized with pDNA (beta-gal) plus pDNA (CCL19). We conclude that CCL19 is an attractive adjuvant for DNA vaccination able to augment antitumor immunity and that this effect is partially caused by enhanced CD8+ T-cell recruitment.


Subject(s)
Antineoplastic Agents/immunology , Chemokines, CC/immunology , Dendritic Cells/drug effects , Neoplasms/therapy , T-Lymphocytes/drug effects , Vaccines, DNA/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Chemokine CCL19 , Chemokines, CC/administration & dosage , Dendritic Cells/immunology , Disease Models, Animal , Mice , Mice, Inbred C57BL , Neoplasms/immunology , T-Lymphocytes/immunology , Th1 Cells/immunology , Th1 Cells/metabolism , Vaccination , Vaccines, DNA/administration & dosage
16.
Pulm Pharmacol Ther ; 19(6): 391-6, 2006.
Article in English | MEDLINE | ID: mdl-16448831

ABSTRACT

Eotaxin is a chemokine that has high potency and selectivity as a chemoattractant agent for eosinophils, signalling exclusively through the CCR3 receptor. Eotaxin is upregulated in the lungs within 3 h of antigen challenge, levels peak at 6 h in lung tissue and bronchoalveolar (BAL) fluid, and fall within 12 h of exposure. This study aimed to look at the effect(s) of eotaxin inhalation on airway function in guinea pigs, to determine if the expected inflammatory cell (eosinophil) infiltration could induce airway hyperreactivity (AHR) and a bronchoconstrictor response equivalent to the late asthmatic response (LAR) seen after antigen challenge. Animals were sensitised with 100 microg/ml OA with a dose on days 1 and 5. Airway responses to inhaled eotaxin (10 or 20 microg/ml) were determined by whole body plethysmography as the change in specific airway conductance (sGaw). Inhaled histamine (1mM) was used to investigate AHR, and cell influx was determined by BAL. Senitised animals exposed to 10 microg/ml eotaxin did not reveal a bronchoconstrictor response or AHR and cellular infiltration to the lungs was not evident 24 h after exposure. Both sensitised and non-sensitised animals exposed to 20 microg/ml eotaxin however revealed a significant bronchoconstrictor response 6h post-challenge, with reductions in sGaw of -27.0+/-6.6% and -32.3+/-6.8%, respectively. Both groups also displayed a bronchoconstrictor response to inhaled histamine 24h after exposure, indicating AHR, and a significant increase in both total and differential cell counts. Sensitised animals, however, revealed a significant increase in cell influx compared to non-sensitised animals. Nebulised eotaxin can reveal a LAR, AHR to inhaled histamine, and cellular infiltration to the lungs, possibly via the mobilisation of eosinophils from the bone marrow, and their subsequent recruitment to the airways.


Subject(s)
Chemokines, CC/pharmacology , Hypersensitivity/pathology , Hypersensitivity/physiopathology , Inflammation/pathology , Respiratory Physiological Phenomena/drug effects , Administration, Inhalation , Airway Resistance/drug effects , Airway Resistance/physiology , Animals , Bronchial Hyperreactivity/chemically induced , Bronchial Hyperreactivity/physiopathology , Bronchoalveolar Lavage Fluid/cytology , Bronchoconstriction/drug effects , Bronchoconstriction/physiology , Cell Count , Chemokine CCL11 , Chemokines, CC/administration & dosage , Data Interpretation, Statistical , Guinea Pigs , Histamine , Male , Ovalbumin/immunology , Plethysmography, Whole Body
17.
Mol Ther ; 13(1): 194-202, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16112911

ABSTRACT

Tyrosinase-related protein-2 (TRP2) is a weak antigen expressed in murine and human melanomas. Induction of antibody (Ab) response and T-cell immunity toward TRP2 with DNA plasmid vaccines has not been efficient to date. Recent studies have suggested that a chemokine ligand for the CCR7 (CCL21) present on T-cells and dendritic cells is important in activating and regulating immunity. We investigated the effectiveness of CCL21 as an adjuvant with an HVJ anionic liposomal TRP2 DNA (plasmid) vaccine to enhance anti-TRP2 Ab, cytokines, delayed-type hypersensitivity, T-cell responses, and tumor protection against B16 melanoma cells. Induction of anti-TRP2 immunity depended mainly on cell-mediated immunity, which was regulated by timing and route of CCL21 administration with DNA vaccine. The optimum protocol was to administer CCL21 im 24 h before DNA vaccine at the same vaccination site. Two vaccinations (prime/boost) were essential for induction of strong anti-TRP2 cell-mediated immunity. CCL21 administration 3 days before or 24 h after DNA vaccine, simultaneous with DNA vaccine, or at different sites (iv, opposite leg) was not effective. This study demonstrated that CCL21 was an effective adjuvant to enhance TRP2-specific immunity induced by a plasmid DNA cancer vaccine.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Cancer Vaccines/therapeutic use , Chemokines, CC/therapeutic use , Intramolecular Oxidoreductases/immunology , Melanoma, Experimental/therapy , Vaccines, DNA/therapeutic use , Adjuvants, Immunologic/administration & dosage , Animals , Antibody Formation , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Cell Line, Tumor , Chemokine CCL21 , Chemokines, CC/administration & dosage , Chemokines, CC/immunology , Female , Immunity, Cellular , Liposomes , Lymph Nodes/immunology , Lymph Nodes/pathology , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Spleen/immunology , Spleen/pathology , Transplantation, Heterologous , Vaccination , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology
18.
Exp Hematol ; 33(10): 1101-8, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16219532

ABSTRACT

OBJECTIVE: CCL23/Myeloid progenitor inhibitory factor-1 is a human CC chemokine with potent in vitro suppressor effects on both human and murine myeloid progenitor cells. This study concerns in vivo inhibitory effect of CCL23 on production of polymorphonuclear leukocytes (PMNs) and monocytes in the bone marrow and their release into the circulation. METHODS: 5'-Bromo-2'-deoxyuridine (BrdU; 100 mg/kg) was used to label dividing PMNs and monocytes in the bone marrow, and BrdU-labeled cells were followed for 10 days in the circulation and identified using immunocytochemistry. Rabbits were given CCL23 (100 mug/kg, n = 5) or saline (control: n = 5) intravenously daily for 3 days before labeling with BrdU. Turnover of PMNs and monocytes in the bone marrow and their transit times through the bone marrow were calculated. RESULTS: CCL23 treatment tended to prolong transit time of PMN (98.4 +/- 4.3 hours vs 111.2 +/- 3.8 hours, control vs CCL23, p = 0.06) through the bone marrow and decreased the size of the bone marrow mitotic pool of PMN (p < 0.01). CCL23 treatment also prolonged the transit time of monocyte (43.4 +/- 3.1 hours vs 54.2 +/- 1.3 hours, control vs CCL23, p < 0.05) through the bone marrow and decreased turnover and pool size of monocytes in the bone marrow (p < 0.05). CONCLUSION: We conclude that CCL23 suppresses PMN and monocyte progenitors, decreases the pool size and slows their turnover in the bone marrow.


Subject(s)
Bone Marrow Cells/physiology , Chemokines, CC/administration & dosage , Leukopoiesis/drug effects , Neutrophils/physiology , Animals , Antimetabolites, Antineoplastic/administration & dosage , Blood Circulation/drug effects , Blood Circulation/physiology , Bone Marrow Cells/cytology , Bromodeoxyuridine/administration & dosage , Female , Humans , Leukopoiesis/physiology , Mice , Monocytes/cytology , Monocytes/physiology , Neutrophils/cytology , Rabbits , Recombinant Proteins/administration & dosage
19.
J Allergy Clin Immunol ; 116(4): 820-6, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16210056

ABSTRACT

BACKGROUND: Little is known about the mechanisms that regulate the selective recruitment of basophils to sites of allergic inflammation. OBJECTIVE: Here we examine the role of stem cell factor (SCF) in the regulation of basophil function. METHODS: Human basophils were isolated from peripheral blood, and their migration was investigated in chemotaxis assays. Apoptosis was detected by means of annexin V and propidium iodide staining. The expression of cell-surface molecules was measured by means of flow cytometry. RESULTS: SCF amplified the chemotactic responsiveness of human peripheral blood basophils to the chemoattractants eotaxin, monocyte chemotactic protein 2 and macrophage inflammatory protein 1alpha, and C5a, without being chemotactic or chemokinetic by itself. SCF synergized with chemoattractants in causing basophil upregulation of the integrin CD11b, and this effect was inhibited by a c-kit antibody, the tyrosine kinase inhibitor imatinib mesylate (STI-571), and a phosphatidylinositol 3 kinase inhibitor but not by inhibitors of p38 mitogen-activated protein kinase or mitogen-activated protein kinase/extracellular signal-regulated kinase kinase. Basophils bound fluorescence-labeled SCF and expressed its receptor, c-kit, which was markedly upregulated in culture for 24 to 48 hours in the presence of IL-3. Moreover, SCF prolonged basophil survival in concert with IL-3 by delaying apoptosis. These effects of SCF were selective for basophils because chemotaxis and CD11b upregulation of eosinophils or neutrophils were unchanged. CONCLUSION: SCF might be an important selective modulator of basophil function through a phosphatidylinositol 3 kinase-dependent pathway.


Subject(s)
Basophils/drug effects , Integrins/metabolism , Stem Cell Factor/pharmacology , Basophils/cytology , Basophils/physiology , CD11b Antigen/metabolism , Cell Survival/drug effects , Chemokine CCL11 , Chemokine CCL4 , Chemokine CCL8 , Chemokines, CC/administration & dosage , Chemotactic Factors/administration & dosage , Chemotaxis, Leukocyte/drug effects , Complement C5a/administration & dosage , Drug Synergism , Humans , In Vitro Techniques , Interleukin-3/administration & dosage , Macrophage Inflammatory Proteins/administration & dosage , Monocyte Chemoattractant Proteins/administration & dosage , Proto-Oncogene Proteins c-kit/metabolism , Signal Transduction/drug effects , Stem Cell Factor/administration & dosage , Up-Regulation/drug effects
20.
Biol Pharm Bull ; 28(6): 1066-70, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15930746

ABSTRACT

Considerable attention has recently been paid to the application of chemokines to cancer immunotherapy because of their chemotactic affinity for a variety of immune cells and because several chemokines are strongly angiostatic. In the present study, the recombinant adenovirus vectors encoding chemokine CCL19 or XCL1 in an E1 cassette (AdRGD-mCCL19 and AdRGD-mXCL1) were developed. The constructed fiber-mutant adenovirus vector, which contained the integrin-targeting Arg-Gly-Asp (RGD) sequence in the fiber knob, notably enhanced the transfection efficiency to OV-HM ovarian carcinoma cells compared to that induced by conventional adenovirus vector. The results of an in vitro chemotaxis assay for chemokine-encoding vector demonstrated that both AdRGD-mCCL19 and AdRGD-mXCL1 could induce the migration of cells expressing specific chemokine receptors. Of the two chemokine-encoding vectors evaluated in vivo, AdRGD-mCCL19 showed significant tumor-suppressive activity in B6C3F1 mice via transduction into OV-HM cells, whereas XCL1 did not exhibit any notable anti-tumor effects, suggesting that CCL19 may be a candidate for cancer immunotherapy.


Subject(s)
Adenoviridae/genetics , Antineoplastic Agents , Chemokines, CC/genetics , Genetic Vectors/genetics , Ovarian Neoplasms/genetics , Transfection/methods , Animals , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Chemokine CCL19 , Chemokines, CC/administration & dosage , Female , Genetic Vectors/administration & dosage , Humans , Mice , Ovarian Neoplasms/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...