Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Biol Pharm Bull ; 44(10): 1548-1550, 2021.
Article in English | MEDLINE | ID: mdl-34602564

ABSTRACT

The antidepressant effect of eicosapentaenoic acid-derived bioactive lipid, resolvin E1 (RvE1), was examined in a murine model of chronic pain-induced depression using a tail suspension test. Because RvE1 reportedly possesses agonistic activity on a chemerin receptor ChemR23, we also examined the antidepressant effect of chemerin. Two weeks after surgery for unilateral spared nerve injury to prepare neuropathic pain model mice, immobility time was measured in a tail suspension test. Chronic pain significantly increased immobility time, and this depression-like behavior was attenuated by intracerebroventricular injection of RvE1 (1 ng) or chemerin (500 ng). These results demonstrate that RvE1 exerts an antidepressant effect in a murine model of chronic pain-induced depression, which is likely to be via ChemR23. RvE1 and its receptor may be promising targets to develop novel antidepressants.


Subject(s)
Antidepressive Agents/administration & dosage , Chronic Pain/complications , Depression/drug therapy , Eicosapentaenoic Acid/analogs & derivatives , Receptors, Chemokine/agonists , Animals , Chemokines/administration & dosage , Chemokines/metabolism , Chronic Pain/psychology , Depression/etiology , Depression/psychology , Disease Models, Animal , Eicosapentaenoic Acid/administration & dosage , Humans , Injections, Intraventricular , Intercellular Signaling Peptides and Proteins/administration & dosage , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mice , Receptors, Chemokine/metabolism , Signal Transduction/drug effects
2.
Life Sci ; 284: 119897, 2021 Nov 01.
Article in English | MEDLINE | ID: mdl-34450172

ABSTRACT

AIM: Contradiction overwhelms chemerin link to feeding behavior. Neither the chemerin central role on appetite regulation nor its relation to hypothalamic histamine and AMPK is verified. MAIN METHODS: Food intake, body weight and hypothalamic biochemical changes were assessed after a single intra-cerebroventricular or intraperitoneal injection (ip) (1 µg/kg or 16 µg/kg, respectively) or chronic ip administration (8 µg/kg/day) of chemerin for 14 or 28 days. Hypothalamic neurobiochemical changes in chemerin/histamine/AMPK induced by either 8-week high fat diet (HFD) or food restriction were also investigated. To confirm chemerin-histamine crosstalk, these neurobiochemical changes were assessed under settings of H1-receptor agonism and/or antagonism by betahistine and/or olanzapine, respectively for 3 weeks. KEY FINDINGS: Chemerin-injected rats exhibited anorexigenic behavior in both acute and chronic studies that was associated with a decreased AMPK activity in the arcuate nucleus (ARC). However, with long-term administration, chemerin anorexigenic effect gradually ceased. Contrarily to food restriction, 8-week HFD increased ARC expression of chemerin and its receptor CMKLR1, reducing food intake via an interplay of H1-receptors and AMPK activity. Blockage of H1-receptors by olanzapine disrupted chemerin signaling pathway with an increased AMPK activity, augmenting food intake. These changes were reversed to normal by betahistine coadministration. SIGNIFICANCE: Chemerin is an anorexigenic adipokine, whose dysregulation is implicated in diet, and olanzapine-induced obesity through a histamine/AMPK axis in the ARC. Hypothalamic chemerin/CMKLR1 expression is a dynamic time-dependent response to changes in body weight and/or food intake. Targeting chemerin as a novel therapeutic approach against antipsychotic- or diet-induced obesity is worth to be further delineated.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Chemokines/metabolism , Diet , Histamine/metabolism , Hypothalamus/metabolism , Obesity/chemically induced , Obesity/metabolism , Olanzapine/adverse effects , Animals , Arcuate Nucleus of Hypothalamus/drug effects , Arcuate Nucleus of Hypothalamus/metabolism , Betahistine/administration & dosage , Body Weight/drug effects , Caloric Restriction , Chemokines/administration & dosage , Diet, High-Fat , Feeding Behavior/drug effects , Female , Histamine H1 Antagonists/pharmacology , Injections, Intraperitoneal , Rats, Wistar , Receptors, Chemokine/metabolism , Receptors, Histamine H1/metabolism
3.
Reprod Sci ; 28(6): 1659-1670, 2021 06.
Article in English | MEDLINE | ID: mdl-33886116

ABSTRACT

Endometrial receptivity and thickness play an important role in achieving a pregnancy. Intrauterine autologous platelet-rich plasma (PRP) infusion has been used in infertile women with recurrent implantation failure (RIF) and thin endometrial lining thickness (EMT). Literature search was performed in PubMed for studies including in vitro, animal, and human studies as well as in abstracts presented at national conferences. Animal studies demonstrated a decrease in the expression of inflammatory markers and fibrosis, and increased endometrial proliferation rate, increased expression of proliferative genes, and increased pregnancy rates. The in vitro studies showed that PRP was associated with increased stromal and mesenchymal cell proliferation, increased expression of regenerative enzymes, and enhancement in cell migration. In infertile women undergoing assisted reproductive technology, one randomized clinical trial showed that PRP intrauterine infusion improved EMT, implantation rate, and clinical pregnancy rate (CPR) in patients with thin EMT, while 3 other trials involving subjects with RIF showed conflicting results related to CPR. Case series and cohort studies showed conflicting results pertaining to CPR. Data to date suggest that PRP may be beneficial in improving endometrial thickness and endometrial receptivity. However, further large prospective and high-quality trials are needed to assert its effect and to identify the population of patients that would benefit the most.


Subject(s)
Endometrium/physiology , Platelet-Rich Plasma , Uterus/physiology , Adult , Animals , Chemokines/administration & dosage , Cytokines/administration & dosage , Embryo Implantation , Embryo Transfer , Endometrium/anatomy & histology , Endometrium/drug effects , Female , Gynatresia/complications , Humans , Infertility, Female/etiology , Infertility, Female/therapy , Injections , Intercellular Signaling Peptides and Proteins/administration & dosage , Platelet-Rich Plasma/chemistry , Platelet-Rich Plasma/physiology , Pregnancy , Reproductive Techniques, Assisted , Uterus/drug effects
4.
Arterioscler Thromb Vasc Biol ; 41(2): 822-836, 2021 02.
Article in English | MEDLINE | ID: mdl-33327748

ABSTRACT

OBJECTIVE: vMIP-II (viral macrophage inflammatory protein 2)/vCCL2 (viral chemotactic cytokine ligand 2) binds to multiple chemokine receptors, and vMIP-II-based positron emission tomography tracer (64Cu-DOTA-vMIP-II: vMIP-II tracer) accumulates at atherosclerotic lesions in mice. Given that it would be expected to react with multiple chemokine receptors on monocytes and macrophages, we wondered if its accumulation in atherosclerosis lesion-bearing mice might correlate with overall macrophage burden or, alternatively, the pace of monocyte recruitment. Approach and Results: We employed a mouse model of atherosclerosis regression involving adenoassociated virus 8 vector encoding murine Apoe (AAV-mApoE) treatment of Apoe-/- mice where the pace of monocyte recruitment slows before macrophage burden subsequently declines. Accumulation of 64Cu-DOTA-vMIP-II at Apoe-/- plaque sites was strong but declined with AAV-mApoE-induced decline in monocyte recruitment, before macrophage burden reduced. Monocyte depletion indicated that monocytes and macrophages themselves were not the only target of the 64Cu-DOTA-vMIP-II tracer. Using fluorescence-tagged vMIP-II tracer, competitive receptor blocking with CXCR4 antagonists, endothelial-specific Cre-mediated deletion of CXCR4, CXCR4-specific tracer 64Cu-DOTA-FC131, and CXCR4 staining during disease progression and regression, we show endothelial cell expression of CXCR4 is a key target of 64Cu-DOTA-vMIP-II imaging. Expression of CXCR4 was low in nonplaque areas but strongly detected on endothelium of progressing plaques, especially on proliferating endothelium, where vascular permeability was increased and monocyte recruitment was the strongest. CONCLUSIONS: Endothelial injury status of plaques is marked by CXCR4 expression and this injury correlates with the tendency of such plaques to recruit monocytes. Furthermore, our findings suggest positron emission tomography tracers that mark CXCR4 can be used translationally to monitor the state of plaque injury and monocyte recruitment.


Subject(s)
Aorta, Thoracic/diagnostic imaging , Atherosclerosis/diagnostic imaging , Chemokines/administration & dosage , Endothelium, Vascular/diagnostic imaging , Molecular Imaging , Monocytes/metabolism , Organometallic Compounds/administration & dosage , Positron-Emission Tomography , Radiopharmaceuticals/administration & dosage , Receptors, CXCR4/metabolism , Animals , Aorta, Thoracic/immunology , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Atherosclerosis/immunology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Biomarkers/metabolism , Cell Line , Chemokines/pharmacokinetics , Disease Models, Animal , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Injections, Intravenous , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Knockout, ApoE , Monocytes/immunology , Monocytes/pathology , Organometallic Compounds/pharmacokinetics , Plaque, Atherosclerotic , Predictive Value of Tests , Radiopharmaceuticals/pharmacokinetics , Receptors, CXCR4/genetics
5.
Pflugers Arch ; 472(6): 673-681, 2020 06.
Article in English | MEDLINE | ID: mdl-32462328

ABSTRACT

Chemerin is an adipocytokine involved in inflammation and lipid metabolism via G protein-coupled receptor, chemokine-like receptor (CMKLR)1. Since the important nuclei regulating pressure (BP) exist in the brain, we examined the effects of acute intracerebroventricular (i.c.v.) injection of chemerin-9 on systemic BP and explored underlying mechanisms. We examined the effects of acute i.c.v. injection of chemerin-9 (10 nmol/head) on systemic BP by a carotid cannulation method in the control or CMKLR1 small interfering (si) RNA-treated Wistar rats (0.04 nmol, 3 days, i.c.v.). We examined protein expression of CMKLR1 around brain ventricles by Western blotting. We examined the effects of acute i.c.v. injection of chemerin-9 on serum adrenaline by a high performance liquid chromatography. In the control siRNA-treated rats, chemerin-9 significantly increased mean BP, which reached a peak at 2 to 4 min after injection. On the other hand, in the CMKLR1 siRNA-treated rats, chemerin-9 did not affect the mean BP. Protein expression of CMKLR1 specifically in subfornical organ (SFO) and paraventricular nucleus (PVN) from the CMKLR1 siRNA-treated rats decreased compared with the control siRNA-treated rats. In the control siRNA-treated rats, chemerin-9 increased serum adrenaline level. On the other hand, in the CMKLR1 siRNA-treated rats, chemerin-9 did not affect the serum adrenaline level. Further, pretreatment with prazosin, an α-adrenaline receptor blocker, significantly prevented the pressor responses induced by chemerin-9. In summary, we for the first time demonstrated that chemerin-9 stimulates the sympathetic nerves via CMKLR1 perhaps expressed in SFO and PVN, which leads to an increase in systemic BP.


Subject(s)
Blood Pressure/drug effects , Brain/drug effects , Brain/metabolism , Chemokines/administration & dosage , Chemokines/metabolism , Receptors, Chemokine/metabolism , Sympathetic Nervous System/drug effects , Animals , Epinephrine/blood , Inflammation/metabolism , Infusions, Intraventricular , Male , Prazosin/pharmacology , RNA, Small Interfering/metabolism , Rats , Rats, Wistar , Sympathetic Nervous System/metabolism
6.
Pancreatology ; 19(3): 401-408, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30833212

ABSTRACT

OBJECTIVE: Chemerin, an adipokine, works as the chemoattractant for the immune cells. The role of chemerin in the inflammatory reaction is controversial. Chemerin has been shown to aggravate the inflammatory response, but other studies demonstrated its anti-inflammatory influence. This study assessed the effects of chemerin on acute pancreatitis (AP) in vivo and in vitro. METHODS: For in vivo experiments male Wistar rats were used. For in vitro study rat pancreatic AR42J cells were employed. Chemerin (1, 5 or 10 µg/kg) was given to the rats prior to the induction of AP by subcutaneous caerulein infusion (25 µg/kg). For in vitro studies cells were subjected to caerulein (10 nM) with or without chemerin (100 nM). Serum amylase activity was measured by enzymatic method, serum TNFα concentration - by ELISA kit. Western-blot was used to examine cellular proteins. RESULTS: AP was confirmed by histological examination. Chemerin given to AP rats decreased histological manifestations of AP, reduced serum amylase activity and TNFα concentration. In AR42J cells subjected to caerulein with addition of chemerin signal for TNFα was reduced comparing to the cultures treated with caerulein alone. Analysis of the dynamics of nuclear translocation for p50, p65 and Bcl-3 points out to NF-κB attenuation as a mechanism of observed anti-inflammatory action of chemerin. CONCLUSION: Chemerin significantly alleviated severity of AP in the rat, this is possibly due to the inhibition of pro-inflammatory signaling in the pancreatic cells.


Subject(s)
Chemokines/therapeutic use , Intercellular Signaling Peptides and Proteins/therapeutic use , NF-kappa B/metabolism , Pancreatitis/chemically induced , Animals , Cell Line , Ceruletide/toxicity , Chemokines/administration & dosage , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Intercellular Signaling Peptides and Proteins/administration & dosage , Male , Pancreas/cytology , Pancreatitis/drug therapy , Rats , Rats, Wistar
7.
Br J Cancer ; 118(10): 1337-1348, 2018 05.
Article in English | MEDLINE | ID: mdl-29717200

ABSTRACT

BACKGROUND: Chemerin, a known chemoattractant, participates in multiple biological events. However, its role in cancer remains largely unknown. METHODS: Chemerin expression was evaluated by real-time PCR, western blot and immunohistochemistry. Forced expression, RNAi, immunoprecipitation, etc. were used in function and mechanism study. Mouse models of extrahepatic and intrahepatic metastasis were employed to evaluate the therapeutic potential of chemerin. RESULTS: Chemerin expression was significantly downregulated in hepatocellular carcinoma, and associated with poor prognosis of HCC patients. Forced expression of chemerin inhibited in vitro migration, invasion and in vivo metastasis of HCC cells. Administration of chemerin effectively suppressed extrahepatic and intrahepatic metastases of HCC cells, resulting in prolonged survival of tumour-bearing nude mice. Chemerin upregulated expression and phosphatase activity of PTEN by interfering with PTEN-CMKLR1 interaction, leading to weakened ubiquitination of PTEN and decreased p-Akt (Ser473) level, which was responsible for suppressed migration, invasion and metastasis of HCC cells. Positive correlation between chemerin and PTEN, and reverse correlation between chemerin and p-Akt (Ser473) were also observed in HCC clinical samples and intrahepatic mouse model in vivo. CONCLUSIONS: Our study has revealed the suppressive role and therapeutic potential of chemerin in HCC metastasis, providing both a prognostic marker and drug candidate for HCC.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Chemokines/administration & dosage , Intercellular Signaling Peptides and Proteins/administration & dosage , Liver Neoplasms/drug therapy , PTEN Phosphohydrolase/genetics , Receptors, Chemokine/genetics , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Movement , Cell Proliferation , Chemokines/genetics , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Humans , Intercellular Signaling Peptides and Proteins/genetics , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Metastasis , Oncogene Protein v-akt/genetics , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
8.
Int J Mol Sci ; 18(1)2017 Jan 13.
Article in English | MEDLINE | ID: mdl-28098795

ABSTRACT

Angiogenesis is involved in the inflammation and proliferation stages of wound healing, to bring inflammatory cells to the wound and provide a microvascular network to maintain new tissue formation. An excess of inflammation, however, leads to prolonged wound healing and scar formation, often resulting in unfavourable outcomes such as amputation. CC-chemokines play key roles in the promotion of inflammation and inflammatory-driven angiogenesis. Therefore, inhibition of the CC-chemokine class may improve wound healing. We aimed to determine if the broad-spectrum CC-chemokine inhibitor "35K" could accelerate wound healing in vivo in mice. In a murine wound healing model, 35K protein or phosphate buffered saline (PBS, control) were added topically daily to wounds. Cohorts of mice were assessed in the early stages (four days post-wounding) and in the later stages of wound repair (10 and 21 days post-wounding). Topical application of the 35K protein inhibited CC-chemokine expression (CCL5, CCL2) in wounds and caused enhanced blood flow recovery and wound closure in early-mid stage wounds. In addition, 35K promoted neovascularisation in the early stages of wound repair. Furthermore, 35K treated wounds had significantly lower expression of the p65 subunit of NF-κB, a key inflammatory transcription factor, and augmented wound expression of the pro-angiogenic and pro-repair cytokine TGF-ß. These findings show that broad-spectrum CC-chemokine inhibition may be beneficial for the promotion of wound healing.


Subject(s)
Chemokines/pharmacology , Neovascularization, Physiologic/drug effects , Wound Healing/drug effects , Administration, Topical , Animals , Biomarkers/metabolism , Chemokines/administration & dosage , Collagen/metabolism , Inflammation/metabolism , Inflammation/pathology , Macrophages/drug effects , Macrophages/metabolism , Mice, Inbred C57BL , Perfusion , Transforming Growth Factor beta/metabolism , Vascular Endothelial Growth Factor A/metabolism
9.
Vascul Pharmacol ; 88: 30-41, 2017 01.
Article in English | MEDLINE | ID: mdl-27890480

ABSTRACT

Chemerin is an adipokine associated with increased blood pressure, and may link obesity with hypertension. We tested the hypothesis that chemerin-induced contraction of the vasculature occurs via calcium flux in smooth muscle cells. Isometric contraction of rat aortic rings was performed in parallel with calcium kinetics of rat aortic smooth muscle cells to assess the possible signaling pathway. Chemerin-9 (nonapeptide of the chemerin S157 isoform) caused a concentration-dependent contraction of isolated aorta (EC50 100nM) and elicited a concentration-dependent intracellular calcium response (EC50 10nM). Pertussis toxin (Gi inhibitor), verapamil (L-type Ca2+ channel inhibitor), PP1 (Src inhibitor), and Y27632 (Rho kinase inhibitor) reduced both calcium influx and isometric contraction to chemerin-9 but PD098059 (Erk MAPK inhibitor) and U73122 (PLC inhibitor) had little to no effect on either measure of chemerin signaling. Although our primary aim was to examine chemerin signaling, we also highlight differences in the mechanisms of chemerin-9 and recombinant chemerin S157. These data support a chemerin-induced contractile mechanism in vascular smooth muscle that functions through Gi proteins to activate L-type Ca2+ channels, Src, and Rho kinase. There is mounting evidence linking chemerin to hypertension and this mechanism brings us closer to targeting chemerin as a form of therapy.


Subject(s)
Aorta/metabolism , Calcium/metabolism , Chemokines/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Myocytes, Smooth Muscle/metabolism , Animals , Aorta/drug effects , Calcium Channels, L-Type/metabolism , Chemokines/administration & dosage , Dose-Response Relationship, Drug , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Hypertension/physiopathology , Intercellular Signaling Peptides and Proteins/administration & dosage , Male , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , rho-Associated Kinases/metabolism
10.
Nat Commun ; 7: 12528, 2016 08 19.
Article in English | MEDLINE | ID: mdl-27538380

ABSTRACT

Chemotherapy remains a mainstay of cancer treatment but its use is often limited by the development of adverse reactions. Severe loss of body weight (cachexia) is a frequent cause of death in cancer patients and is exacerbated by chemotherapy. We show that genetic inactivation of vascular endothelial growth factor (VEGF)-A in myeloid cells prevents chemotherapy-induced cachexia by inhibiting skeletal muscle loss and the lipolysis of white adipose tissue. It also improves clearance of senescent tumour cells by natural killer cells and inhibits tumour regrowth after chemotherapy. The effects depend on the chemoattractant chemerin, which is released by the tumour endothelium in response to chemotherapy. The findings define chemerin as a critical mediator of the immune response, as well as an important inhibitor of cancer cachexia. Targeting myeloid cell-derived VEGF signalling should impede the lipolysis and weight loss that is frequently associated with chemotherapy, thereby substantially improving the therapeutic outcome.


Subject(s)
Antineoplastic Agents/therapeutic use , Cachexia/drug therapy , Killer Cells, Natural/immunology , Myeloid Cells/immunology , Neoplasms/drug therapy , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Animals , Antineoplastic Agents/pharmacology , Cachexia/etiology , Cachexia/immunology , Cachexia/pathology , Chemokines/administration & dosage , Chemokines/immunology , Humans , Intercellular Signaling Peptides and Proteins/administration & dosage , Intercellular Signaling Peptides and Proteins/immunology , Lipolysis/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Neoplasms/complications , Neoplasms/immunology , Neoplasms/pathology , Treatment Outcome , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
11.
Sci Rep ; 6: 26830, 2016 05 26.
Article in English | MEDLINE | ID: mdl-27225311

ABSTRACT

Long-term and reversible changes in body weight are typical of seasonal animals. Thyroid hormone (TH) and retinoic acid (RA) within the tanycytes and ependymal cells of the hypothalamus have been implicated in the photoperiodic response. We investigated signalling downstream of RA and how this links to the control of body weight and food intake in photoperiodic F344 rats. Chemerin, an inflammatory chemokine, with a known role in energy metabolism, was identified as a target of RA. Gene expression of chemerin (Rarres2) and its receptors were localised within the tanycytes and ependymal cells, with higher expression under long (LD) versus short (SD) photoperiod, pointing to a physiological role. The SD to LD transition (increased food intake) was mimicked by 2 weeks of ICV infusion of chemerin into rats. Chemerin also increased expression of the cytoskeletal protein vimentin, implicating hypothalamic remodelling in this response. By contrast, acute ICV bolus injection of chemerin on a 12 h:12 h photoperiod inhibited food intake and decreased body weight with associated changes in hypothalamic neuropeptides involved in growth and feeding after 24 hr. We describe the hypothalamic ventricular zone as a key site of neuroendocrine regulation, where the inflammatory signal, chemerin, links TH and RA signaling to hypothalamic remodeling.


Subject(s)
Chemokines/physiology , Energy Metabolism/physiology , Hypothalamus/metabolism , Intercellular Signaling Peptides and Proteins/physiology , Photoperiod , Animals , Body Weight/drug effects , Chemokines/administration & dosage , Chemokines/pharmacology , Eating/drug effects , Energy Metabolism/drug effects , Ependyma/cytology , Ependyma/metabolism , Ependymoglial Cells/metabolism , Humans , Hypothalamus/cytology , Hypothalamus/drug effects , Injections, Intraventricular , Intercellular Signaling Peptides and Proteins/administration & dosage , Intercellular Signaling Peptides and Proteins/pharmacology , Male , Neuronal Plasticity/drug effects , Random Allocation , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Receptors, Chemokine/analysis , Receptors, Chemokine/drug effects , Signal Transduction/drug effects , Thyroid Hormones/physiology
12.
Nanomedicine (Lond) ; 11(6): 693-714, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27003586

ABSTRACT

Cardiovascular diseases make up one of the main causes of death today, with myocardial infarction and ischemic heart disease contributing a large share of the deaths reported. With mainstream clinical therapy focusing on palliative medicine following myocardial infarction, the structural changes that occur in the diseased heart will eventually lead to end-stage heart failure. Heart transplantation remains the only gold standard of cure but a shortage in donor organs pose a major problem that led to clinicians and researchers looking into alternative strategies for cardiac repair. This review will examine some alternative methods of treatment using chemokines and drugs carried by nanoparticles as drug delivering agents for the purposes of treating myocardial infarction through the promotion of revascularization. We will also provide an overview of existing studies involving such nanoparticulate drug delivery systems, their reported efficacy and the challenges facing their translation into ubiquitous clinical use.


Subject(s)
Drug Delivery Systems/methods , Heart/drug effects , Myocardial Infarction/drug therapy , Nanoparticles/chemistry , Angiopoietins/administration & dosage , Angiopoietins/therapeutic use , Animals , Chemokines/administration & dosage , Chemokines/therapeutic use , Fibroblast Growth Factors/administration & dosage , Fibroblast Growth Factors/therapeutic use , Humans , Neovascularization, Physiologic/drug effects , Vascular Endothelial Growth Factor A/administration & dosage , Vascular Endothelial Growth Factor A/therapeutic use
13.
Mucosal Immunol ; 9(1): 13-23, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25943275

ABSTRACT

There have been encouraging results for the development of an effective HIV vaccine. However, many questions remain regarding the quality of immune responses and the role of mucosal antibodies. We addressed some of these issues by using a simian immunodeficiency virus (SIV) DNA vaccine adjuvanted with plasmid-expressed mucosal chemokines combined with an intravaginal SIV challenge in rhesus macaque (RhM) model. We previously reported on the ability of CCR9 and CCR10 ligand (L) adjuvants to enhance mucosal and systemic IgA and IgG responses in small animals. In this study, RhMs were intramuscularly immunized five times with either DNA or DNA plus chemokine adjuvant delivered by electroporation followed by challenge with SIVsmE660. Sixty-eight percent of all vaccinated animals (P<0.01) remained either uninfected or had aborted infection compared with only 14% in the vaccine naïve group. The highest protection was observed in the CCR10L chemokines group, where six of nine animals had aborted infection and two remained uninfected, leading to 89% protection (P<0.001). The induction of mucosal SIV-specific antibodies and neutralization titers correlated with trends in protection. These results indicate the need to further investigate the contribution of chemokine adjuvants to modulate immune responses and the role of mucosal antibodies in SIV/HIV protection.


Subject(s)
AIDS Vaccines/administration & dosage , Antibodies, Viral/biosynthesis , Chemokines/immunology , Immunity, Mucosal/drug effects , Simian Acquired Immunodeficiency Syndrome/prevention & control , Vaccines, DNA/administration & dosage , AIDS Vaccines/genetics , AIDS Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Chemokines/administration & dosage , Chemokines/genetics , Female , Immunity, Cellular/drug effects , Ligands , Macaca mulatta , Plasmids/chemistry , Plasmids/immunology , Receptors, CCR/genetics , Receptors, CCR/immunology , Receptors, CCR10/genetics , Receptors, CCR10/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/drug effects , Simian Immunodeficiency Virus/immunology , Vaccination , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Vagina/drug effects , Vagina/immunology , Vagina/virology
14.
World J Gastroenterol ; 21(14): 4184-94, 2015 Apr 14.
Article in English | MEDLINE | ID: mdl-25892868

ABSTRACT

AIM: To test whether hepatic stellate cells (HSCs) at different activation stages play different roles in acetaminophen (APAP)-induced acute liver injury (ALI). METHODS: HSCs were isolated from mouse liver and cultured in vitro. Morphological changes of initiation HSCs [HSCs (5d)] and perpetuation HSCs [HSCs (p3)] were observed by immunofluorescence and transmission electron microscopy. The protective effects of HSC-derived molecules, cell lysates and HSC-conditioned medium (HSC-CM) were tested in vivo by survival and histopathological analyses. Liver injury was determined by measuring aminotransferase levels in the serum and by histologic examination of tissue sections under a light microscope. Additionally, to determine the molecular mediators of the observed protective effects of initiation HSCs, we examined HSC-CM using a high-density protein array. RESULTS: HSCs (5d) and HSCs (p3) had different morphological and phenotypic traits. HSCs (5d) presented a star-shaped appearance with expressing α-SMA at non-uniform levels between cells. However, HSCs (p3) evolved into myofibroblast-like cells without lipid droplets and expressed a uniform and higher level of α-SMA. HSC-CM (5d), but not HSC-CM (p3), provided a significant survival benefit and showed a dramatic reduction of hepatocellular necrosis and panlobular leukocyte infiltrates in mice exposed to APAP. However, this protective effect was abrogated at higher cell masses, indicating a therapeutic window of effectiveness. Furthermore, the protein array screen revealed that HSC-CM (5d) was composed of many chemokines and growth factors that correlated with inflammatory inhibition and therapeutic activity. When compared with HSC-CM (p3), higher levels of monocyte chemoattractant protein-1, macrophage inflammatory protein-1γ, hepatocyte growth factor, interleukin-10, and matrix metalloproteinase-2, but lower levels of stem cell factor and Fas-Ligand were observed in HSC-CM (5d). CONCLUSION: These data indicated that initiation HSCs and perpetuation HSCs were different in morphology and protein expression, and provided the first experimental evidence of the potential medical value of initiation HSC-derived molecules in the treatment of ALI.


Subject(s)
Chemical and Drug Induced Liver Injury/metabolism , Chemokines/metabolism , Hepatic Stellate Cells/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Liver/metabolism , Paracrine Communication , Acetaminophen , Animals , Anti-Inflammatory Agents/administration & dosage , Cell Shape , Cells, Cultured , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/pathology , Chemokines/administration & dosage , Culture Media, Conditioned/metabolism , Disease Models, Animal , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/ultrastructure , Intercellular Signaling Peptides and Proteins/administration & dosage , Liver/drug effects , Liver/ultrastructure , Male , Mice, Inbred C57BL , Necrosis , Phenotype , Signal Transduction , Time Factors
15.
Gen Comp Endocrinol ; 196: 34-40, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24287340

ABSTRACT

The effects of intraperitoneal injections of cholecystokinin (CCK), apelin, ghrelin, and orexin on food intake were examined in the blind cavefish Astyanax fasciatus mexicanus. CCK (50ng/g) induced a decrease in food intake whereas apelin (100ng/g), orexin (100ng/g), and ghrelin (100ng/g) induced an increase in food intake as compared to saline-injected control fish. In order to better understand the central mechanism by which these hormones act, we examined the effects of injections on the brain mRNA expression of two metabolic enzymes, tyrosine hydroxylase (TH), and mechanistic target of rapamycin (mTOR), and of appetite-regulating peptides, CCK, orexin, apelin and cocaine and amphetamine regulated transcript (CART). CCK injections induced a decrease in brain apelin injections, apelin injections induced an increase in TH, mTOR, and orexin brain expressions, orexin treatment increased brain TH expression and ghrelin injections induced an increase in mTOR and orexin brain expressions. CART expression was not affected by any of the injection treatments. Our results suggest that the enzymes TH and mTOR and the hormones CCK, apelin, orexin, and ghrelin all regulate food intake in cavefish through a complex network of interactions.


Subject(s)
Appetite/physiology , Brain/drug effects , Cholecystokinin/administration & dosage , Eating/drug effects , Ghrelin/administration & dosage , Hormones/pharmacology , Intracellular Signaling Peptides and Proteins/administration & dosage , Neuropeptides/administration & dosage , Sirolimus/pharmacology , Tyrosine 3-Monooxygenase/metabolism , Animals , Brain/enzymology , Characidae , Chemokines/administration & dosage , Cholagogues and Choleretics/administration & dosage , Immunosuppressive Agents/pharmacology , Neurotransmitter Agents/administration & dosage , Orexins , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Tyrosine 3-Monooxygenase/antagonists & inhibitors , Tyrosine 3-Monooxygenase/genetics
16.
CNS Neurol Disord Drug Targets ; 12(6): 815-29, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24047524

ABSTRACT

Chemokines may play a role in leukocyte migration across the blood-brain barrier (BBB) during neuroinflammation and other neuropathological processes, such as epilepsy. The CC chemokine receptor 5 (CCR5) is a member of CC-chemokine receptor family that binds several chemokines, including CCL3 (macrophage inflammatory protein-1alpha, MIP-1alpha), CCL4 (macrophage inflammatory protein-1beta, MIP-1beta) and CCL5 (RANTES). The current review examines the relationship between CCR5 and the microglia in different neurological disorders and models of CNS injury. CCR5 expression is upregulated in different neurological diseases, where it is often immunolocalized in microglial cells. A multistep cascade couples CCR5 activation by chemokines to Ca(2+) increases in human microglia. Because changes in [Ca(2+)] (i) affect chemotaxis, secretion, and gene expression, pharmacologic modulation of this pathway may alter inflammatory and degenerative processes in the CNS. Consequently, targeting CCR5 by using CCR5 antagonists may attenuate critical aspects of neuroinflammation in different models of neurological disorders. To illustrate the interaction between CCR5 and microglia in the CNS, we used a model of excitotoxicity, and demonstrate the intimate involvement of CCR5 in neuron injury and inflammation attendant to kainic acid (KA)-induced neurotoxicity. CCR5 participates in neuronal injury caused by the excitotoxin, KA, brings inflammatory cells to the sites of KA-induced CNS injury, defines the extent of tissue loss after KA exposure and limits reparative responses. We used a SV40-derived vector carrying an interfering RNA (RNAi) that targets CCR5. Delivered directly to the bone marrow, this vector decreased CCR5 expression in circulating cells. Animals so treated showed greatly reduced expression of CCR5 and its ligands (MIP-1alpha and RANTES) in the CNS, including in the brain vasculature, decreased BBB leakage, demonstrated greater KA-stimulated neurogenesis and increased migration of bone marrow-derived cells to the brain to become neurons. Thus, therapeutic targeting of CCR5 may allow control of potentially injurious neuroinflammatory responses, including decrease in microglial cells activation and proliferation, and facilitate neurogenic repair in seizure-induced and, potentially, other forms of CNS injury.


Subject(s)
Disease Models, Animal , Drug Delivery Systems/methods , Epilepsy/metabolism , Inflammation/metabolism , Inflammation/pathology , Microglia/metabolism , Nerve Regeneration/physiology , Receptors, CCR5/metabolism , Animals , CCR5 Receptor Antagonists , Cell Death/drug effects , Cell Death/physiology , Chemokines/administration & dosage , Chemokines/metabolism , Epilepsy/drug therapy , Epilepsy/pathology , Humans , Inflammation/drug therapy , Microglia/drug effects , Nerve Regeneration/drug effects , Nervous System Diseases/drug therapy , Nervous System Diseases/metabolism , Nervous System Diseases/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology
17.
Blood ; 120(7): 1449-57, 2012 Aug 16.
Article in English | MEDLINE | ID: mdl-22618707

ABSTRACT

The chemokine CCL3/MIP-1α is a risk factor in the outcome of multiple myeloma (MM), particularly in the development of osteolytic bone disease. This chemokine, highly overexpressed by MM cells, can signal mainly through 2 receptors, CCR1 and CCR5, only 1 of which (CCR1) is responsive to CCL3 in human and mouse osteoclast precursors. CCR1 activation leads to the formation of osteolytic lesions and facilitates tumor growth. Here we show that formation of mature osteoclasts is blocked by the highly potent and selective CCR1 antagonist CCX721, an analog of the clinical compound CCX354. We also show that doses of CCX721 selected to completely inhibit CCR1 produce a profound decrease in tumor burden and osteolytic damage in the murine 5TGM1 model of MM bone disease. Similar effects were observed when the antagonist was used prophylactically or therapeutically, with comparable efficacy to that of zoledronic acid. 5TGM1 cells were shown to express minimal levels of CCR1 while secreting high levels of CCL3, suggesting that the therapeutic effects of CCX721 result from CCR1 inhibition on non-MM cells, most likely osteoclasts and osteoclast precursors. These results provide a strong rationale for further development of CCR1 antagonists for the treatment of MM and associated osteolytic bone disease.


Subject(s)
Chemokines/pharmacology , Chemokines/therapeutic use , Multiple Myeloma/drug therapy , Osteolysis/drug therapy , Receptors, CCR1/antagonists & inhibitors , Tumor Burden/drug effects , Administration, Oral , Animals , Cell Death/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cellular Microenvironment/drug effects , Chemokines/administration & dosage , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Immunocompetence/drug effects , Inflammation/drug therapy , Inflammation/pathology , Mice , Mice, Inbred C57BL , Models, Biological , Monocytes/drug effects , Monocytes/metabolism , Multiple Myeloma/complications , Multiple Myeloma/pathology , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoclasts/pathology , Osteolysis/complications , Osteolysis/pathology , Rats , Receptors, CCR1/metabolism
18.
Peptides ; 32(9): 1866-71, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21855588

ABSTRACT

Visceral adipose tissue-derived serpin (vaspin) improves glucose tolerance and insulin sensitivity in diet-induced obese mice. Chemerin may increase insulin sensitivity in adipose tissue and seems to be associated with several key aspects of metabolic syndrome. Decreased levels of omentin-1 are associated with increasing obesity and insulin resistance. Our study aimed to investigate the effects of vaspin, chemerin and omentin-1 acute administration on feeding and hypothalamic gene expression of peptides which play a key role in feeding regulation. 35 rats were injected into the arcuate nucleus (ARC) of the hypothalamus with either saline (n=8), vaspin (1µg/kg; n=9), chemerin (8µg/kg; n=9), or omentin-1 (8µg/kg; n=9). Food intake in the following 24h was recorded, thereafter rats were sacrificed. Total RNA was extracted from hypothalami and reverse transcribed to evaluate hypothalamic gene expression of agouti-related peptide (AgRP), neuropeptide Y (NPY), orexin-A, cocaine- and amphetamine-regulated transcript (CART), corticotrophin releasing hormone (CRH) and proopiomelanocortin (POMC), by real-time reverse transcription polymerase chain reaction. Compared to vehicle, vaspin injection significantly decreased feeding, while chemerin and omentin-1 had no effect in the tested dose. Vaspin treatment significantly decreased NPY and increased POMC gene expression. Chemerin treatment led to a significant increase of both AgRP and POMC gene expression. Omentin-1 treatment did not modify gene expression of the investigated peptides. Therefore, vaspin is an adipokine triggering anorectic pathways in the hypothalamus, where reduction of NPY and increase of POMC mRNA levels mediate feeding inhibition. Chemerin and omentin-1 have no effect on feeding in the tested dose.


Subject(s)
Chemokines/pharmacology , Cytokines/pharmacology , Feeding Behavior/drug effects , Hypothalamus/drug effects , Lectins/pharmacology , Serpins/pharmacology , Agouti-Related Protein/genetics , Agouti-Related Protein/metabolism , Animals , Appetite Depressants/pharmacology , Chemokines/administration & dosage , Cytokines/administration & dosage , Eating/drug effects , GPI-Linked Proteins/administration & dosage , GPI-Linked Proteins/pharmacology , Gene Expression Regulation , Humans , Hypothalamus/cytology , Intercellular Signaling Peptides and Proteins , Lectins/administration & dosage , Male , Mice , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Neuropeptides/genetics , Neuropeptides/metabolism , Polymerase Chain Reaction/methods , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/metabolism , Rats , Rats, Wistar , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Serpins/administration & dosage
19.
J Control Release ; 155(2): 184-92, 2011 Oct 30.
Article in English | MEDLINE | ID: mdl-21708196

ABSTRACT

Patients with malignant non-Hodgkin's lymphomas (NHL) of B-cell lineages relapse despite initial anti-tumor response to chemotherapy or antibody treatments. Failure to eliminate the tumor is often because of inadequate priming, low cell numbers and suboptimal phenotype of effector T cells. Here we describe a new biomaterial-based controlled-release paradigm to treat weakly immunogenic NHLs by in-situ amplifying the number of functional, antigen-specific T-helper 1 (Th1) cells following immunotherapy. An injectable, synthetic immune priming center (sIPC) consisting of an in-situ crosslinking, chemokine-carrying hydrogel and both DNA- and siRNA dual-loaded microparticles, is reported. This sIPC chemo attracts a large number of immature dendritic cells (DCs) at the site of administration and efficiently co-delivers both DNA antigens and interleukin-10 (IL10)-silencing siRNA to those cells. Using a murine model of A20 B cell lymphoma, we demonstrate that combination of DNA-antigen delivery and IL10 silencing, synergistically activate recruited immature DCs and cause a strong shift towards Th1 response while suppressing Th2 and Th17 cytokines. sIPC-based immunotherapy showed 45% more CD8+ cytotoxic T cell (CTL) response and 53% stronger CD4+ CTL activity compared to naked DNA vaccine. In addition, in-vivo sIPC immunization induced significant protection (p<0.01) against subsequent tumor challenge. Such a multi-modal, injectable system that simultaneously delivers chemokines, siRNA and DNA antigens to DCs marks a new approach to in-situ priming and modulation during immunotherapy and could provide effective vaccination strategies against cancers and infectious diseases.


Subject(s)
Cancer Vaccines/administration & dosage , Chemokines/administration & dosage , DNA/administration & dosage , Drug Carriers/chemistry , Lymphoma, B-Cell/immunology , RNA, Small Interfering/administration & dosage , Th1 Cells , Animals , Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Chemokines/immunology , Cross-Linking Reagents , DNA/immunology , Delayed-Action Preparations , Dendritic Cells/cytology , Dendritic Cells/immunology , Female , Hydrogels , Immunoglobulin Idiotypes/immunology , Injections, Intramuscular , Interleukin-10/immunology , Lymphoma, B-Cell/therapy , Mice , Mice, Inbred BALB C , RNA, Small Interfering/immunology , T-Lymphocytes, Cytotoxic/immunology , Th1 Cells/cytology , Th1 Cells/immunology
20.
J Immunol ; 186(4): 2219-28, 2011 02 15.
Article in English | MEDLINE | ID: mdl-21242526

ABSTRACT

Exosomes derived from dendritic cells or tumor cells are a population of nanometer-sized membrane vesicles that can induce specific antitumor immunity. During investigation of the effects of hyperthermia on antitumor immune response, we found that exosomes derived from heat-stressed tumor cells (HS-TEX) could chemoattract and activate dendritic cells (DC) and T cells more potently than that by conventional tumor-derived exosomes. We show that HS-TEX contain chemokines, such as CCL2, CCL3, CCL4, CCL5, and CCL20, and the chemokine-containing HS-TEX are functionally competent in chemoattracting CD11c(+) DC and CD4(+)/CD8(+) T cells both in vitro and in vivo. Moreover, the production of chemokine-containing HS-TEX could be inhibited by ATP inhibitor, calcium chelator, and cholesterol scavenger, indicating that the mobilization of chemokines into exosomes was ATP- and calcium-dependent and via a lipid raft-dependent pathway. We consistently found that the intracellular chemokines could be enriched in lipid rafts after heat stress. Accordingly, intratumoral injection of HS-TEX could induce specific antitumor immune response more efficiently than that by tumor-derived exosomes, thus inhibiting tumor growth and prolonging survival of tumor-bearing mice more significantly. Therefore, our results demonstrate that exosomes derived from HS-TEX represent a kind of efficient tumor vaccine and can chemoattract and activate DC and T cells, inducing more potent antitumor immune response. Release of chemokines through exosomes via lipid raft-dependent pathway may be a new method of chemokine exocytosis.


Subject(s)
Cancer Vaccines/immunology , Carcinoma, Lewis Lung/prevention & control , Chemokines/metabolism , Exosomes/metabolism , Heat Stress Disorders/prevention & control , Melanoma, Experimental/prevention & control , Membrane Microdomains/immunology , Signal Transduction/immunology , Animals , Cancer Vaccines/administration & dosage , Cancer Vaccines/metabolism , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/metabolism , Cell Line, Tumor , Cells, Cultured , Chemokines/administration & dosage , Chemokines/immunology , Coculture Techniques , Dendritic Cells/immunology , Dendritic Cells/metabolism , Exosomes/immunology , Heat Stress Disorders/immunology , Heat Stress Disorders/metabolism , Hot Temperature , Male , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Membrane Microdomains/metabolism , Mice , Mice, Inbred C57BL , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...