Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Semin Immunol ; 59: 101603, 2022 01.
Article in English | MEDLINE | ID: mdl-35341670

ABSTRACT

Melanocortin receptors have emerged as important targets with a very unusual versatility, as their widespread distribution on multiple tissues (e.g. skin, adrenal glands, brain, immune cells, exocrine glands) together with the variety of physiological processes they control (pigmentation, cortisol release, satiety mechanism, inflammation, secretions), place this family of receptors as genuine therapeutic targets for many disorders. This review focuses in the journey of the development of melanocortin receptors as therapeutic targets from the discovery of their existence in the early 1990 s to the approval of the first few drugs of this class. Two major areas of development characterise the current state of melanocortin drug development: their role in obesity, recently culminated with the approval of setmelanotide, and their potential for the treatment of chronic inflammatory and autoimmune diseases like rheumatoid arthritis, multiple sclerosis or fibrosis. The pro-resolving nature of these drugs offers the advantage of acting by mimicking the way our body naturally resolves inflammation, expecting fewer side effects and a more balanced (i.e. non-immunosuppressive) response from them. Here we also review the approaches followed for the design and development of novel compounds, the importance of the GPCR nature of these receptors in the process of drug development, therapeutic value, current challenges and successes, and the potential for the implementation of precision medicine approaches through the incorporation of genetics advances.


Subject(s)
Arthritis, Rheumatoid , Melanocortins , Humans , Carrier Proteins , Inflammation/drug therapy , Inflammation/metabolism , Melanocortins/metabolism , Receptors, Melanocortin/genetics , Receptors, Melanocortin/metabolism
2.
Cell Mol Life Sci ; 78(7): 3333-3354, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33439271

ABSTRACT

In recent years, cellular senescence has become the focus of attention in multiple areas of biomedical research. Typically defined as an irreversible cell cycle arrest accompanied by increased cellular growth, metabolic activity and by a characteristic messaging secretome, cellular senescence can impact on multiple physiological and pathological processes such as wound healing, fibrosis, cancer and ageing. These unjustly called 'zombie cells' are indeed a rich source of opportunities for innovative therapeutic development. In this review, we collate the current understanding of the process of cellular senescence and its two-faced nature, i.e. beneficial/detrimental, and reason this duality is linked to contextual aspects. We propose the senescence programme as an endogenous pro-resolving mechanism that may lead to sustained inflammation and damage when dysregulated or when senescent cells are not cleared efficiently. This pro-resolving model reconciles the paradoxical two faces of senescence by emphasising that it is the unsuccessful completion of the programme, and not senescence itself, what leads to pathology. Thus, pro-senescence therapies under the right context, may favour inflammation resolution. We also review the evidence for the multiple therapeutic approaches under development based on senescence, including its induction, prevention, clearance and the use of senolytic and senomorphic drugs. In particular, we highlight the importance of the immune system in the favourable outcome of senescence and the implications of an inefficient immune surveillance in completion of the senescent cycle. Finally, we identify and discuss a number of challenges and existing gaps to encourage and stimulate further research in this exciting and unravelled field, with the hope of promoting and accelerating the clinical success of senescence-based therapies.


Subject(s)
Aging , Cellular Senescence , Fibrosis/pathology , Immune System , Neoplasms/pathology , Wound Healing , Animals , Cell Proliferation , Humans , Translational Research, Biomedical
3.
J Immunol ; 198(7): 2876-2885, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28242648

ABSTRACT

We hypothesized that exudates collected at the beginning of the resolution phase of inflammation might be enriched for tissue protective molecules; thus an integrated cellular and molecular approach was applied to identify novel chondroprotective bioactions. Exudates were collected 6 h (inflammatory) and 24 h (resolving) following carrageenan-induced pleurisy in rats. The resolving exudate was subjected to gel filtration chromatography followed by proteomics, identifying 61 proteins. Fractions were added to C28/I2 chondrocytes, grown in micromasses, ions with or without IL-1ß or osteoarthritic synovial fluids for 48 h. Three proteins were selected from the proteomic analysis, α1-antitrypsin (AAT), hemopexin (HX), and gelsolin (GSN), and tested against catabolic stimulation for their effects on glycosaminoglycan deposition as assessed by Alcian blue staining, and gene expression of key anabolic proteins by real-time PCR. In an in vivo model of inflammatory arthritis, cartilage integrity was determined histologically 48 h after intra-articular injection of AAT or GSN. The resolving exudate displayed protective activities on chondrocytes, using multiple readouts: these effects were retained in low m.w. fractions of the exudate (46.7% increase in glycosaminoglycan deposition; ∼20% upregulation of COL2A1 and aggrecan mRNA expression), which reversed the effect of IL-1ß. Exogenous administration of HX, GSN, or AAT abrogated the effects of IL-1ß and osteoarthritic synovial fluids on anabolic gene expression and increased glycosaminoglycan deposition. Intra-articular injection of AAT or GSN protected cartilage integrity in mice with inflammatory arthritis. In summary, the strategy for identification of novel chondroprotective activities in resolving exudates identified HX, GSN and AAT as potential leads for new drug discovery programs.


Subject(s)
Arthritis, Experimental/pathology , Chondrocytes/drug effects , Exudates and Transudates/chemistry , Pleurisy/immunology , Animals , Disease Models, Animal , Male , Mass Spectrometry , Mice , Mice, Inbred C57BL , Osteoarthritis, Knee/pathology , Proteomics , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction
4.
Semin Immunol ; 27(3): 216-26, 2015 May.
Article in English | MEDLINE | ID: mdl-25726511

ABSTRACT

Although anti-inflammatory drugs are among the most common class of marketed drugs, chronic inflammatory conditions such as rheumatoid arthritis, multiple sclerosis or inflammatory bowel disease still represent unmet needs. New first-in-class drugs might be discovered in the future but the repurpose and further development of old drugs also offers promise for these conditions. This is the case of the melanocortin adrenocorticotropin hormone, ACTH, used in patients since 1952 but regarded as the last therapeutic option when other medications, such as glucocorticoids, cannot be used. Better understanding on its physiological and pharmacological mechanisms of actions and new insights on melanocortin receptors biology have revived the interest on rescuing this old and effective drug. ACTH does not only induce cortisol production, as previously assumed, but it also exerts anti-inflammatory actions by targeting melanocortin receptors present on immune cells. The endogenous agonists for these receptors (ACTH, α-, ß-, and γ-melanocyte stimulating hormones), are also produced locally by immune cells, indicating the existence of an endogenous anti-inflammatory tissue-protective circuit involving the melanocortin system. These findings suggested that new ACTH-like melanocortin drugs devoid of steroidogenic actions, and hence side effects, could be developed. This review summarizes the actions of ACTH and melanocortin drugs, their role as endogenous pro-resolving mediators, their current clinical use and provides an overview on how recent advances on GPCR functioning may lead to a novel class of drugs.


Subject(s)
Adrenocorticotropic Hormone/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Inflammation/drug therapy , Receptors, Melanocortin/agonists , Amino Acid Sequence , Arthritis, Rheumatoid/drug therapy , Homeostasis/immunology , Humans , Inflammation/immunology , Inflammation/pathology , Inflammatory Bowel Diseases/drug therapy , Molecular Sequence Data , Multiple Sclerosis/drug therapy , Receptors, Melanocortin/metabolism , Signal Transduction/immunology
5.
PLoS Biol ; 13(7): e1002202, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26181372

ABSTRACT

Lactate has long been considered a "waste" by-product of cell metabolism, and it accumulates at sites of inflammation. Recent findings have identified lactate as an active metabolite in cell signalling, although its effects on immune cells during inflammation are largely unexplored. Here we ask whether lactate is responsible for T cells remaining entrapped in inflammatory sites, where they perpetuate the chronic inflammatory process. We show that lactate accumulates in the synovia of rheumatoid arthritis patients. Extracellular sodium lactate and lactic acid inhibit the motility of CD4+ and CD8+ T cells, respectively. This selective control of T cell motility is mediated via subtype-specific transporters (Slc5a12 and Slc16a1) that we find selectively expressed by CD4+ and CD8+ subsets, respectively. We further show both in vitro and in vivo that the sodium lactate-mediated inhibition of CD4+ T cell motility is due to an interference with glycolysis activated upon engagement of the chemokine receptor CXCR3 with the chemokine CXCL10. In contrast, we find the lactic acid effect on CD8+ T cell motility to be independent of glycolysis control. In CD4+ T helper cells, sodium lactate also induces a switch towards the Th17 subset that produces large amounts of the proinflammatory cytokine IL-17, whereas in CD8+ T cells, lactic acid causes the loss of their cytolytic function. We further show that the expression of lactate transporters correlates with the clinical T cell score in the synovia of rheumatoid arthritis patients. Finally, pharmacological or antibody-mediated blockade of subtype-specific lactate transporters on T cells results in their release from the inflammatory site in an in vivo model of peritonitis. By establishing a novel role of lactate in control of proinflammatory T cell motility and effector functions, our findings provide a potential molecular mechanism for T cell entrapment and functional changes in inflammatory sites that drive chronic inflammation and offer targeted therapeutic interventions for the treatment of chronic inflammatory disorders.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , Inflammation/metabolism , Lactic Acid/metabolism , Animals , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Cell Movement , Chemokines/metabolism , Female , Glycolysis , Humans , Inflammation/immunology , Mice, Inbred C57BL , Synovial Fluid/metabolism
6.
Cell Mol Life Sci ; 74(7): 1335-1345, 2017 04.
Article in English | MEDLINE | ID: mdl-27853832

ABSTRACT

The efficiency of drug research and development has paradoxically declined over the last decades despite major scientific and technological advances, promoting new cost-effective strategies such as drug repositioning by systematic screening for new actions of known drugs. Here, we performed a screening for positive allosteric modulators (PAMs) at melanocortin (MC) receptors. The non-steroidal anti-inflammatory drug fenoprofen, but not the similar compound ibuprofen, presented PAM activity at MC3, MC4, and MC5 receptors. In a model of inflammatory arthritis, fenoprofen afforded potent inhibition while ibuprofen was nearly inactive. Fenoprofen presented anti-arthritic actions on cartilage integrity and synovitis, effects markedly attenuated in Mc3r-/- mice. Fenoprofen displayed pro-resolving properties promoting macrophage phagocytosis and efferocytosis, independently of cyclooxygenase inhibition. In conclusion, combining repositioning with advances in G-protein coupled receptor biology (allosterism) may lead to potential new therapeutics. In addition, MC3 PAMs emerged as a viable approach to the development of innovative therapeutics for joint diseases.


Subject(s)
Allosteric Regulation/drug effects , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Drug Repositioning , Fenoprofen/pharmacology , Receptor, Melanocortin, Type 3/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Arthritis/drug therapy , Arthritis/etiology , CHO Cells , Cricetinae , Cricetulus , Disease Models, Animal , Fenoprofen/therapeutic use , Joints/metabolism , Joints/pathology , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Male , Melanocortins/analysis , Mice , Mice, Inbred C57BL , Mice, Knockout , Peritonitis/chemically induced , Peritonitis/drug therapy , Peritonitis/pathology , Phagocytosis/drug effects , Prostaglandin-Endoperoxide Synthases/chemistry , Prostaglandin-Endoperoxide Synthases/metabolism , Receptor, Melanocortin, Type 3/chemistry , Receptor, Melanocortin, Type 3/deficiency , Receptor, Melanocortin, Type 3/genetics
7.
FASEB J ; 30(12): 4033-4041, 2016 12.
Article in English | MEDLINE | ID: mdl-27535487

ABSTRACT

Alveolar bone loss is a result of an aggressive form of periodontal disease (PD) associated with Aggregatibacter actinomycetemcomitans (Aa) infection. PD is often observed with other systemic inflammatory conditions, including arthritis. Melanocortin peptides activate specific receptors to exert antiarthritic properties, avoiding excessing inflammation and modulating macrophage function. Recent work has indicated that melanocortin can control osteoclast development and function, but whether such protection takes place in infection-induced alveolar bone loss has not been investigated. The purpose of this study was to evaluate the role of melanocortin in Aa-induced PD. Mice were orally infected with Aa and treated with the melanocortin analog DTrp8-γMSH or vehicle daily for 30 d. Then, periodontal tissue was collected and analyzed. Aa-infected mice treated with DTrp8-γMSH presented decreased alveolar bone loss and a lower degree of neutrophil infiltration in the periodontium than vehicle-treated animals; these actions were associated with reduced periodontal levels of TNF-α, IFN-γ, and IL-17A. In vitro experiments with cells differentiated into osteoclasts showed that osteoclast formation and resorptive activity were attenuated after treatment with DTrp8-γMSH. Thus, melanocortin agonism could represent an innovative way to tame overexuberant inflammation and, at the same time, preserve bone physiology, as seen after Aa infection.-Madeira, M. F. M., Queiroz-Junior, C. M., Montero-Melendez, T., Werneck, S. M. C., Corrêa, J. D., Soriani, F. M., Garlet, G. P., Souza, D. G., Teixeira, M. M., Silva, T. A., Perretti, M. Melanocortin agonism as a viable strategy to control alveolar bone loss induced by oral infection.


Subject(s)
Alveolar Bone Loss/prevention & control , Melanocortins/agonists , Osteoclasts/microbiology , Pasteurellaceae Infections/prevention & control , Periodontal Diseases/metabolism , Aggregatibacter actinomycetemcomitans , Alveolar Bone Loss/etiology , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Macrophages/immunology , Mice, Inbred C57BL , Periodontitis/drug therapy , Periodontitis/metabolism
8.
J Immunol ; 194(7): 3381-8, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25725103

ABSTRACT

There is a need for novel approaches to control pathologies with overexuberant inflammatory reactions. Targeting melanocortin (MC) receptors represents a promising therapy for obesity and chronic inflammation, but lack of selectivity and safety concerns limit development. A new way to increase selectivity of biological effects entails the identification of biased agonists. In this study, we characterize the small molecule AP1189 as a biased agonist at receptors MC1 and MC3. Although not provoking canonical cAMP generation, AP1189 addition to MC1 or MC3, but not empty vector, transfected HEK293 cells caused ERK1/2 phosphorylation, a signaling responsible for the proefferocytic effect evoked in mouse primary macrophages. Added to macrophage cultures, AP1189 reduced cytokine release, an effect reliant on both MC1 and MC3 as evident from the use of Mc1r(-/-) and Mc3r(-/-) macrophages. No melanogenesis was induced by AP1189 in B16-F10 melanocytes. In vivo, oral AP1189 elicited anti-inflammatory actions in peritonitis and, upon administration at the peak of inflammation, accelerated the resolution phase by ∼3-fold. Finally, given the clinical efficacy of adrenocorticotropin in joint diseases, AP1189 was tested in experimental inflammatory arthritis, where this biased agonist afforded significant reduction of macroscopic and histological parameters of joint disruption. These proof-of-concept analyses with AP1189, an active oral anti-inflammatory and resolution-promoting compound, indicate that biased agonism at MC receptors is an innovative, viable approach to yield novel anti-inflammatory molecules endowed with a more favorable safety profile.


Subject(s)
Guanidines/pharmacology , Pyrroles/pharmacology , Receptors, Melanocortin/agonists , Receptors, Melanocortin/metabolism , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacology , Arthritis, Experimental/drug therapy , Arthritis, Experimental/genetics , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Calcium/metabolism , Disease Models, Animal , Gene Knockout Techniques , Guanidines/administration & dosage , HEK293 Cells , Humans , Inflammation/drug therapy , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Male , Melanins/metabolism , Melanoma, Experimental , Mice , Neutrophils/immunology , Neutrophils/metabolism , Peritonitis/chemically induced , Peritonitis/drug therapy , Peritonitis/genetics , Peritonitis/metabolism , Peritonitis/pathology , Phagocytosis/immunology , Pyrroles/administration & dosage , Receptor, Melanocortin, Type 1/agonists , Receptor, Melanocortin, Type 1/genetics , Receptor, Melanocortin, Type 1/metabolism , Receptor, Melanocortin, Type 3/agonists , Receptor, Melanocortin, Type 3/genetics , Receptor, Melanocortin, Type 3/metabolism , Receptors, Melanocortin/genetics , Signal Transduction/drug effects
9.
Proc Natl Acad Sci U S A ; 110(45): 18232-7, 2013 Nov 05.
Article in English | MEDLINE | ID: mdl-24108355

ABSTRACT

Formyl-peptide receptor type 2 (FPR2), also called ALX (the lipoxin A4 receptor), conveys the proresolving properties of lipoxin A4 and annexin A1 (AnxA1) and the proinflammatory signals elicited by serum amyloid protein A and cathelicidins, among others. We tested here the hypothesis that ALX might exist as homo- or heterodimer with FPR1 or FPR3 (the two other family members) and operate in a ligand-biased fashion. Coimmunoprecipitation and bioluminescence resonance energy transfer assays with transfected HEK293 cells revealed constitutive dimerization of the receptors; significantly, AnxA1, but not serum amyloid protein A, could activate ALX homodimers. A p38/MAPK-activated protein kinase/heat shock protein 27 signaling signature was unveiled after AnxA1 application, leading to generation of IL-10, as measured in vitro (in primary monocytes) and in vivo (after i.p. injection in the mouse). The latter response was absent in mice lacking the ALX ortholog. Using a similar approach, ALX/FPR1 heterodimerization evoked using the panagonist peptide Ac2-26, identified a JNK-mediated proapoptotic path that was confirmed in primary neutrophils. These findings provide a molecular mechanism that accounts for the dual nature of ALX and indicate that agonist binding and dimerization state contribute to the conformational landscape of FPRs.


Subject(s)
Annexin A1/metabolism , Protein Conformation , Receptors, Formyl Peptide/chemistry , Receptors, Formyl Peptide/metabolism , Signal Transduction/physiology , Amino Acid Sequence , Animals , Bioluminescence Resonance Energy Transfer Techniques , Dimerization , HEK293 Cells , Humans , Immunoprecipitation , Interleukin-10/metabolism , Mice , Molecular Sequence Data , Serum Amyloid A Protein/metabolism
10.
Am J Pathol ; 184(8): 2333-41, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24979595

ABSTRACT

Because there is clinical evidence for an association between periodontal disease and rheumatoid arthritis, it is important to develop suitable experimental models to explore pathogenic mechanisms and therapeutic opportunities. The K/BxN serum model of inflammatory arthritis was applied using distinct protocols, and modulation of joint disruption afforded by dexamethasone and calcitonin was established in comparison to the melanocortin (MC) receptor agonist DTrp(8)-γ-melanocyte stimulating hormone (MSH; DTrp). Wild-type and MC receptor type 3 (MC3)-null mice of different ages were also used. There was significant association between severity of joint disease, induced with distinct protocols and volumes of the arthritogenic K/BxN serum, and periodontal bone damage. Therapeutic treatment with 10 µg dexamethasone, 30 ng elcatonin, and 20 µg DTrp per mouse revealed unique and distinctive pharmacological properties, with only DTrp protecting both joint and periodontal tissue. Further analyses in nonarthritic animals revealed higher susceptibility to periodontal bone loss in Mc3r(-/-) compared with wild-type mice, with significant exacerbation at 14 weeks of age. These data reveal novel protective properties of endogenous MC3 on periodontal status in health and disease and indicate that MC3 activation could lead to the development of a new genus of anti-arthritic bone-sparing therapeutics.


Subject(s)
Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/metabolism , Periodontal Diseases/metabolism , Receptor, Melanocortin, Type 3/metabolism , Animals , Arthritis, Experimental/complications , Arthritis, Rheumatoid/complications , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Periodontal Diseases/complications
11.
Mol Cell Proteomics ; 12(8): 2205-19, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23660474

ABSTRACT

Altered plasma neutrophil microparticle levels have recently been implicated in a number of vascular and inflammatory diseases, yet our understanding of their actions is very limited. Herein, we investigate the proteome of neutrophil microparticles in order to shed light on their biological actions. Stimulation of human neutrophils, either in suspension or adherent to an endothelial monolayer, led to the production of microparticles containing >400 distinct proteins with only 223 being shared by the two subsets. For instance, postadherent microparticles were enriched in alpha-2 macroglobulin and ceruloplasmin, whereas microparticles produced by neutrophils in suspension were abundant in heat shock 70 kDa protein 1. Annexin A1 and lactotransferrin were expressed in both microparticle subsets. We next determined relative abundance of these proteins in three types of human microparticle samples: healthy volunteer plasma, plasma of septic patients and skin blister exudates finding that these proteins were differentially expressed on neutrophil microparticles from these samples reflecting in part the expression profiles we found in vitro. Functional assessment of the neutrophil microparticles subsets demonstrated that in response to direct stimulation neutrophil microparticles produced reactive oxygen species and leukotriene B4 as well as locomoted toward a chemotactic gradient. Finally, we investigated the actions of the two neutrophil microparticles subsets described herein on target cell responses. Microarray analysis with human primary endothelial cells incubated with either microparticle subset revealed a discrete modulation of endothelial cell gene expression profile. These findings demonstrate that neutrophil microparticles are heterogenous and can deliver packaged information propagating the activation status of the parent cell, potentially exerting novel and fundamental roles both under homeostatic and disease conditions.


Subject(s)
Cell-Derived Microparticles/metabolism , Neutrophils/physiology , Adult , Cells, Cultured , Chemotaxis, Leukocyte , Female , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Leukotriene B4/metabolism , Male , Middle Aged , Proteome , Reactive Oxygen Species/metabolism
12.
Br J Nutr ; 109(3): 394-401, 2013 Feb 14.
Article in English | MEDLINE | ID: mdl-22717234

ABSTRACT

Plant-based whole foods provide thousands of bioactive metabolites to the human diet that reduce the risk of developing chronic diseases. ß-Caryophyllene (CAR) is a common constituent of the essential oil of numerous plants, vegetables, fruits and medicinal herbs, and has been used as a flavouring agent since the 1930 s. Here, we report the antioxidant activity of CAR, its protective effect on liver fibrosis and its inhibitory capacity on hepatic stellate cell (HSC) activation. CAR was tested for the inhibition of lipid peroxidation and as a free radical scavenger. CAR had higher inhibitory capacity on lipid peroxidation than probucol, α-humulene and α-tocopherol. Also, CAR showed high scavenging activities against hydroxyl radical and superoxide anion. The activity of 5-lipoxygenase, an enzyme that actively participates in fibrogenesis, was significantly inhibited by CAR. Carbon tetrachloride-treated rats received CAR at 2, 20 and 200 mg/kg. CAR significantly improved liver structure, and reduced fibrosis and the expression of Col1a1, Tgfb1 and Timp1 genes. Oxidative stress was used to establish a model of HSC activation with overproduction of extracellular matrix proteins. CAR (1 and 10 µm) increased cell viability and significantly reduced the expression of fibrotic marker genes. CAR, a sesquiterpene present in numerous plants and foods, is as a natural antioxidant that reduces carbon tetrachloride-mediated liver fibrosis and inhibits hepatic cell activation.


Subject(s)
Antioxidants/therapeutic use , Carbon Tetrachloride Poisoning/prevention & control , Dietary Supplements , Hepatic Stellate Cells/metabolism , Liver/metabolism , Sesquiterpenes/therapeutic use , Animals , Antioxidants/administration & dosage , Antioxidants/metabolism , Antioxidants/pharmacology , Arachidonate 5-Lipoxygenase/chemistry , Arachidonate 5-Lipoxygenase/metabolism , Carbon Tetrachloride Poisoning/metabolism , Carbon Tetrachloride Poisoning/pathology , Cell Line , Cell Survival/drug effects , Female , Flavoring Agents/administration & dosage , Flavoring Agents/metabolism , Flavoring Agents/therapeutic use , Free Radical Scavengers/administration & dosage , Free Radical Scavengers/metabolism , Free Radical Scavengers/therapeutic use , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/enzymology , Lipid Peroxidation/drug effects , Lipoxygenase Inhibitors/administration & dosage , Lipoxygenase Inhibitors/metabolism , Lipoxygenase Inhibitors/therapeutic use , Liver/drug effects , Liver/pathology , Monocyclic Sesquiterpenes , Oxidative Stress/drug effects , Polycyclic Sesquiterpenes , Probucol/pharmacology , Probucol/therapeutic use , Random Allocation , Rats , Rats, Wistar , Sesquiterpenes/administration & dosage , Sesquiterpenes/metabolism , alpha-Tocopherol/metabolism , alpha-Tocopherol/therapeutic use
13.
Ann Rheum Dis ; 71(1): 143-9, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21998117

ABSTRACT

OBJECTIVE: To facilitate the targeting to inflammation sites of small anti-inflammatory peptides, with short half-lives, by fusion with the latency-associated peptide (LAP) of transforming growth factor ß1 through a cleavable matrix metalloproteinase (MMP) linker. This design improves efficacy, overcoming the limitations to their clinical use. METHODS: We generated latent forms of vasoactive intestinal peptide (VIP), α-melanocyte-stimulating hormone (MSH) and γ(3)MSH by fusion to LAP through an MMP cleavage site using recombinant DNA technology. The biological activities of these latent therapeutics were studied in vivo using monosodium urate (MSU)-induced peritonitis and collagen-induced arthritis (CIA) models. We assessed gene therapy and purified protein therapy. RESULTS: The recruitment of the polymorphonuclear cells induced by MSU injection into mouse peritoneal cavity was reduced by 35% with γ(3)MSH (1 nmol), whereas administration of a much lower dose of purified latent LAP-MMP-γ(3)MSH (0.03 nmol) attenuated leucocyte influx by 50%. Intramuscular gene delivery of plasmids coding LAP-MMP-VIP and LAP-MMP-αMSH at disease onset reduced the development of CIA compared with LAP-MMP, which does not contain any therapeutic moiety. Histological analysis confirmed a significantly lower degree of inflammation, bone and cartilage erosion in groups treated with LAP-MMP-VIP or LAP-MMP-αMSH. Antibody titres to collagen type II and inflammatory cytokine production were also reduced in these two groups. CONCLUSION: Incorporation of small anti-inflammatory peptides within the LAP shell and delivered as recombinant protein or through gene therapy can control inflammatory and arthritic disease. This platform delivery can be developed to control human arthritides and other autoimmune diseases.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Arthritis, Experimental/therapy , Melanocyte-Stimulating Hormones/therapeutic use , Peptide Fragments/therapeutic use , Peritonitis/therapy , Recombinant Fusion Proteins/therapeutic use , Transforming Growth Factor beta/therapeutic use , Vasoactive Intestinal Peptide/therapeutic use , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Arthritis, Experimental/drug therapy , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Cytokines/blood , Drug Delivery Systems , Drug Design , Drug Evaluation, Preclinical/methods , Genetic Therapy/methods , Half-Life , Male , Melanocyte-Stimulating Hormones/genetics , Melanocyte-Stimulating Hormones/pharmacokinetics , Mice , Mice, Inbred DBA , Peptide Fragments/genetics , Peptide Fragments/pharmacokinetics , Peritonitis/drug therapy , Peritonitis/metabolism , Recombinant Fusion Proteins/pharmacokinetics , Tissue Distribution , Treatment Outcome , Vasoactive Intestinal Peptide/genetics , Vasoactive Intestinal Peptide/pharmacokinetics
14.
Am J Pathol ; 179(1): 259-69, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21703408

ABSTRACT

Synthetic and natural melanocortin (MC) peptides afford inhibitory properties in inflammation and tissue injury, but characterization of receptor involvement is still elusive. We used the agonist AP214 to test MC-dependent anti-inflammatory effects. In zymosan peritonitis, treatment of mice with AP214 (400 to 800 µg/kg) inhibited cell infiltration, an effect retained in MC receptor type 1, or MC(1), mutant mice but lost in MC(3) null mice. In vitro, cytokine release from zymosan-stimulated macrophages was affected by AP214, with approximately 80%, 30%, and 40% reduction in IL-1ß, tumor necrosis factor-α, and IL-6, respectively. Inhibition of IL-1ß release was retained in MC(1) mutant cells but was lost in MC(3) null cells. Furthermore, AP214 augmented uptake of zymosan particles and human apoptotic neutrophils by wild-type macrophages: this proresolving property was lost in MC(3) null macrophages. AP214 displayed its pro-efferocytotic effect also in vivo. Finally, in a model of inflammatory arthritis, AP214 evoked significant reductions in the clinical score. These results indicate that AP214 elicits anti-inflammatory responses, with a preferential effect on IL-1ß release. Furthermore, we describe for the first time a positive modulation of an MC agonist on the process of efferocytosis. In all cases, endogenous MC(3) is the receptor that mediates these novel properties of AP214. These findings might clarify the tissue-protective properties of AP214 in clinical settings and may open further development for novel MC agonists.


Subject(s)
Arthritis, Experimental/drug therapy , Inflammation/drug therapy , Melanocortins/agonists , Peritonitis/drug therapy , Receptor, Melanocortin, Type 1/physiology , Receptor, Melanocortin, Type 3/physiology , alpha-MSH/analogs & derivatives , Animals , Apoptosis/drug effects , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Blotting, Western , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Humans , Inflammation/metabolism , Inflammation/pathology , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Neutrophils/cytology , Neutrophils/drug effects , Neutrophils/metabolism , Peritonitis/metabolism , Peritonitis/pathology , Phagocytosis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , alpha-MSH/pharmacology
15.
Front Immunol ; 13: 1084394, 2022.
Article in English | MEDLINE | ID: mdl-36793548

ABSTRACT

Stromal cells have emerged as central drivers in multiple and diverse diseases, and consequently, as potential new cellular targets for the development of novel therapeutic strategies. In this review we revise the main roles of fibroblasts, not only as structural cells but also as players and regulators of immune responses. Important aspects like fibroblast heterogeneity, functional specialization and cellular plasticity are also discussed as well as the implications that these aspects may have in disease and in the design of novel therapeutics. An extensive revision of the actions of fibroblasts on different conditions uncovers the existence of numerous diseases in which this cell type plays a pathogenic role, either due to an exacerbation of their 'structural' side, or a dysregulation of their 'immune side'. In both cases, opportunities for the development of innovative therapeutic approaches exist. In this regard, here we revise the existing evidence pointing at the melanocortin pathway as a potential new strategy for the treatment and management of diseases mediated by aberrantly activated fibroblasts, including scleroderma or rheumatoid arthritis. This evidence derives from studies involving models of in vitro primary fibroblasts, in vivo models of disease as well as ongoing human clinical trials. Melanocortin drugs, which are pro-resolving mediators, have shown ability to reduce collagen deposition, activation of myofibroblasts, reduction of pro-inflammatory mediators and reduced scar formation. Here we also discuss existing challenges, both in approaching fibroblasts as therapeutic targets, and in the development of novel melanocortin drug candidates, that may help advance the field and deliver new medicines for the management of diseases with high medical needs.


Subject(s)
Arthritis, Rheumatoid , Melanocortins , Humans , Melanocortins/metabolism , Melanocortins/therapeutic use , Fibroblasts/metabolism , Myofibroblasts/metabolism , Collagen/metabolism , Arthritis, Rheumatoid/metabolism
16.
Front Immunol ; 13: 1078678, 2022.
Article in English | MEDLINE | ID: mdl-36505403

ABSTRACT

Background: Melanocortins are peptides endowed with anti-inflammatory and pro-resolving activities. Many of these effects are mediated by the Melanocortin receptor 1 (MC1) as reported in several experimental settings. As such, MC1 can be a viable target for the development of new therapies that mimic endogenous pro-resolving mediators. The aim of this study was to assess the immunopharmacology of a selective MC1 agonist (PL8177) in vitro and in a mouse model of inflammatory arthritis. Methods: PL8177 and the natural agonist αMSH were tested for activation of mouse and human Melanocortin receptors (MC1,3,4,5), monitoring cAMP accumulation and ERK1/2 phosphorylation, using transiently transfected HEK293A cells. The anti-inflammatory and pro-resolving effects of PL8177 and αMSH were evaluated using mouse peritoneal Macrophages. Finally, a model of K/BxN serum transfer induced arthritis was used to determine the in vivo potential of PL8177. Results: PL8177 activates mouse and human MC1 with apparent EC50 values of 0.01 and 1.49 nM, respectively, using the cAMP accumulation assay. Similar profiles were observed for the induction of ERK phosphorylation (EC50: 0.05 and 1.39 nM). PL8177 displays pro-resolving activity (enhanced Macrophage efferocytosis) and counteracts the inflammatory profile of zymosan-stimulated macrophages, reducing the release of IL-1ß, IL-6, TNF-α and CCL-2. In the context of joint inflammation, PL8177 (3mg/kg i.p.) reduces clinical score, paw swelling and incidence of severe disease as well as the recruitment of immune cells into the arthritic joint. Conclusion: These results demonstrate that the MC1 agonism with PL8177 affords therapeutic effects in inflammatory conditions including arthritis. Significance: Drugs targeting the Melanocortin system have emerged as promising therapeutics for several conditions including inflammation or obesity. Multiple candidates are under clinical development, and some have already reached approval. Here we present the characterization of a novel drug candidate, PL8177, selective for the Melanocortin 1 receptor (MC1), demonstrating its selectivity profile on cAMP and ERK1/2 phosphorylation signaling pathways, of relevance as selective drugs will translate into lesser off-target effect. PL8177 also demonstrated, not only anti-inflammatory activity, but pro-resolving actions due to its ability to enhance efferocytosis (i.e. the phagocytosis of apoptotic cells), endowing this molecule with therapeutic advantages compared to classical anti-inflammatory drugs. Using a mouse model of inflammatory arthritis, the compound demonstrated in vivo efficacy by reducing clinical score, paw swelling and overall disease severity. Taken together, these results present Melanocortin-based therapies, and specifically targeting MC1 receptor, as a promising strategy to manage chronic inflammatory diseases.


Subject(s)
Arthritis , Phagocytosis , Humans , Arthritis/drug therapy , alpha-MSH , Inflammation/drug therapy , Macrophages
17.
Curr Rheumatol Rep ; 13(2): 138-45, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21243457

ABSTRACT

Gouty arthritis is a form of acute joint inflammation provoked by joint deposition of urate crystals. Although this acute pathology resolves after a few days, the marked degree of inflammation in the joint and--possibly more important to the patient--the excruciating pain it causes require proper therapeutic management. Often deemed a "poor sibling" of chronic joint pathologies such as rheumatoid arthritis and psoriatic arthritis, the increasing incidence of gout makes it a more palatable disease for novel drug discovery programs. This fact, associated with novel insights into the molecular mechanisms activated by the urate crystal deposition, is at the basis of new therapeutics under clinical development for gout, a valid example being the effective targeting of the proinflammatory cytokine interleukin-1. Here we briefly review the current status of antigout drug development and propose another target; our focus is on melanocortin receptor agonists as novel therapeutics for gout and inflammatory arthritides, a prototype of which, the adrenocorticotropic hormone, is already used in clinical settings.


Subject(s)
Arthritis, Gouty/drug therapy , Gout Suppressants/therapeutic use , Melanocortins/metabolism , Receptors, Melanocortin/metabolism , Allopurinol/therapeutic use , Animals , Arthritis, Gouty/metabolism , Colchicine/therapeutic use , Crystallization , Disease Models, Animal , Humans , Injections, Intra-Articular , Melanocortins/pharmacology , Mice , Receptors, Melanocortin/drug effects , Uric Acid/administration & dosage , Uric Acid/metabolism
18.
Cell Death Differ ; 28(11): 3140-3160, 2021 11.
Article in English | MEDLINE | ID: mdl-34040168

ABSTRACT

Regulatory T-cells (Tregs) are central in the maintenance of homeostasis and resolution of inflammation. However, the mechanisms that govern their differentiation and function are not completely understood. Herein, we demonstrate a central role for the lipid mediator biosynthetic enzyme 15-lipoxygenase (ALOX15) in regulating key aspects of Treg biology. Pharmacological inhibition or genetic deletion of ALOX15 in Tregs decreased FOXP3 expression, altered Treg transcriptional profile and shifted their metabolism. This was linked with an impaired ability of Alox15-deficient cells to exert their pro-resolving actions, including a decrease in their ability to upregulate macrophage efferocytosis and a downregulation of interferon gamma expression in Th1 cells. Incubation of Tregs with the ALOX15-derived specilized pro-resolving mediators (SPM)s Resolvin (Rv)D3 and RvD5n-3 DPA rescued FOXP3 expression in cells where ALOX15 activity was inhibited. In vivo, deletion of Alox15 led to increased vascular lipid load and expansion of Th1 cells in mice fed western diet, a phenomenon that was reversed when Alox15-deficient mice were reconstituted with wild type Tregs. Taken together these findings demonstrate a central role of pro-resolving lipid mediators in governing the differentiation of naive T-cells to Tregs.


Subject(s)
Arachidonate 15-Lipoxygenase/metabolism , T-Lymphocytes, Regulatory/metabolism , Animals , Cell Differentiation , Healthy Volunteers , Humans , Male , Mice , Up-Regulation
19.
J Extracell Vesicles ; 10(6): 12084, 2021 04.
Article in English | MEDLINE | ID: mdl-33936566

ABSTRACT

Extracellular vesicles (EVs) are emerging as key players in different stages of atherosclerosis. Here we provide evidence that EVs released by mixed aggregates of monocytes and platelets in response to TNF-α display pro-inflammatory actions on endothelial cells and atherosclerotic plaques. Tempering platelet activation with Iloprost, Aspirin or a P2Y12 inhibitor impacted quantity and phenotype of EV produced. Proteomics of EVs from cells activated with TNF-α alone or in the presence of Iloprost revealed a distinct composition, with interesting hits like annexin-A1 and gelsolin. When added to human atherosclerotic plaque explants, EVs from TNF-α stimulated monocytes augmented release of cytokines. In contrast, EVs generated by TNF-α together with Iloprost produced minimal plaque activation. Notably, patients with coronary artery disease that required percutaneous coronary intervention had elevated plasma numbers of monocyte, platelet as well as double positive EV subsets. In conclusion, EVs released following monocyte/platelet activation may play a potential role in the development and progression of atherosclerosis. Whereas attenuating platelet activation modifies EV composition released from monocyte/platelet aggregates, curbing their pro-inflammatory actions may offer therapeutic avenues for the treatment of atherosclerosis.


Subject(s)
Extracellular Vesicles/physiology , Monocytes/physiology , Plaque, Atherosclerotic/physiopathology , Platelet Aggregation/physiology , Aspirin/pharmacology , Atherosclerosis/physiopathology , Blood Platelets/cytology , Blood Platelets/drug effects , Cytokines , Endothelial Cells/drug effects , Extracellular Vesicles/drug effects , Extracellular Vesicles/metabolism , Healthy Volunteers , Humans , Inflammation/immunology , Monocytes/cytology , Platelet Activation/drug effects , Tumor Necrosis Factor-alpha
20.
Nat Commun ; 11(1): 745, 2020 02 06.
Article in English | MEDLINE | ID: mdl-32029712

ABSTRACT

Rheumatoid arthritis affects individuals commonly during the most productive years of adulthood. Poor response rates and high costs associated with treatment mandate the search for new therapies. Here we show that targeting a specific G-protein coupled receptor promotes senescence in synovial fibroblasts, enabling amelioration of joint inflammation. Following activation of the melanocortin type 1 receptor (MC1), synovial fibroblasts acquire a senescence phenotype characterized by arrested proliferation, metabolic re-programming and marked gene alteration resembling the remodeling phase of wound healing, with increased matrix metalloproteinase expression and reduced collagen production. This biological response is attained by selective agonism of MC1, not shared by non-selective ligands, and dependent on downstream ERK1/2 phosphorylation. In vivo, activation of MC1 leads to anti-arthritic effects associated with induction of senescence in the synovial tissue and cartilage protection. Altogether, selective activation of MC1 is a viable strategy to induce cellular senescence, affording a distinct way to control joint inflammation and arthritis.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Receptor, Melanocortin, Type 1/agonists , Animals , Arthritis, Experimental/drug therapy , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Cells, Cultured , Cellular Senescence/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Genetic Variation , Humans , Imidazoles/pharmacology , Male , Metabolic Networks and Pathways/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Melanocortin, Type 1/deficiency , Receptor, Melanocortin, Type 1/genetics , Receptors, Notch/antagonists & inhibitors , Synovial Membrane/drug effects , Synovial Membrane/metabolism , Synovial Membrane/pathology , alpha-MSH/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL