Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Thiago M. Cunha is active.

Publication


Featured researches published by Thiago M. Cunha.


Journal of Leukocyte Biology | 2008

Crucial role of neutrophils in the development of mechanical inflammatory hypernociception

Thiago M. Cunha; Waldiceu A. Verri; Ieda R. S. Schivo; Marcelo Henrique Napimoga; Carlos Amílcar Parada; Stephen Poole; Mauro M. Teixeira; Sérgio H. Ferreira; Fernando Q. Cunha

Neutrophil migration is responsible for tissue damage observed in inflammatory diseases. Neutrophils are also implicated in inflammatory nociception, but mechanisms of their participation have not been elucidated. In the present study, we addressed these mechanisms in the carrageenan‐induced mechanical hypernociception, which was determined using a modification of the Randall‐Sellito test in rats. Neutrophil accumulation into the plantar tissue was determined by the contents of myeloperoxidase activity, whereas cytokines and PGE2 levels were measured by ELISA and radioimmunoassay, respectively. The pretreatment of rats with fucoidin (a leukocyte adhesion inhibitor) inhibited carrageenan‐induced hypernociception in a dose‐ and time‐dependent manner. Inhibition of hypernociception by fucoidin was associated with prevention of neutrophil recruitment, as it did not inhibit the hypernociception induced by the direct‐acting hypernociceptive mediators, PGE2 and dopamine, which cause hypernociception, independent of neutrophils. Fucoidin had no effect on carrageenan‐induced TNF‐α, IL‐1β, and cytokine‐induced neutrophil chemoattractant 1 (CINC‐1)/CXCL1 production, suggesting that neutrophils were not the source of hypernociceptive cytokines. Conversely, hypernociception and neutrophil migration induced by TNF‐α, IL‐1β, and CINC‐1/CXCL1 was inhibited by fucoidin, suggesting that neutrophils are involved in the production of direct‐acting hypernociceptive mediators. Indeed, neutrophils stimulated in vitro with IL‐1β produced PGE2, and IL‐1β‐induced PGE2 production in the rat paw was inhibited by the pretreatment with fucoidin. In conclusion, during the inflammatory process, the migrating neutrophils participate in the cascade of events leading to mechanical hypernociception, at least by mediating the release of direct‐acting hypernociceptive mediators, such as PGE2. Therefore, the blockade of neutrophil migration could be a target to development of new analgesic drugs.


Annals of the Rheumatic Diseases | 2010

IL-33 induces neutrophil migration in rheumatoid arthritis and is a target of anti-TNF therapy

Waldiceu A. Verri; Fabricio O. Souto; Silvio M. Vieira; Sérgio C. L. de Almeida; Sandra Y. Fukada; Damo Xu; José C. Alves-Filho; Thiago M. Cunha; Ana T. Guerrero; Rafaela B Mattos-Guimaraes; Fabíola Reis Oliveira; Mauro M. Teixeira; João S. Silva; Iain B. McInnes; Sérgio H. Ferreira; Paulo Louzada-Junior; Foo Y. Liew; Fernando Q. Cunha

Objectives Interleukin 33 (IL-33) is a new member of the IL-1 family of cytokines which signals via its receptor, ST2 (IL-33R), and has an important role in Th2 and mast cell responses. This study shows that IL-33 orchestrates neutrophil migration in arthritis. Methods and results Methylated bovine serum albumin (mBSA) challenge in the knee joint of mBSA-immunised mice induced local neutrophil migration accompanied by increased IL-33R and IL-33 mRNA expression. Cell migration was inhibited by systemic and local treatments with soluble (s)IL-33R, an IL-33 decoy receptor, and was not evident in IL-33R-deficient mice. IL-33 injection also induced IL-33R-dependent neutrophil migration. Antigen- and IL-33-induced neutrophil migration in the joint was dependent on CXCL1, CCL3, tumour necrosis factor α (TNFα) and IL-1β synthesis. Synovial tissue, macrophages and activated neutrophils expressed IL-33R. IL-33 induces neutrophil migration by activating macrophages to produce chemokines and cytokines and by directly acting on neutrophils. Importantly, neutrophils from patients with rheumatoid arthritis successfully treated with anti-TNFα antibody (infliximab) expressed significantly lower levels of IL-33R than patients treated with methotrexate alone. Only neutrophils from patients treated with methotrexate alone or from normal donors stimulated with TNFα responded to IL-33 in chemotaxis. Conclusions These results suggest that suppression of IL-33R expression in neutrophils, preventing IL-33-induced neutrophil migration, may be an important mechanism of anti-TNFα therapy of inflammation.


Proceedings of the National Academy of Sciences of the United States of America | 2008

IL-33 mediates antigen-induced cutaneous and articular hypernociception in mice

Waldiceu A. Verri; Ana T. Guerrero; Sandra Y. Fukada; Daniel Augusto Rodrigues Valério; Thiago M. Cunha; Damo Xu; Sérgio H. Ferreira; Foo Y. Liew; Fernando Q. Cunha

IL-33, a new member of the IL-1 family, signals through its receptor ST2 and induces T helper 2 (Th2) cytokine synthesis and mediates inflammatory response. We have investigated the role of IL-33 in antigen-induced hypernociception. Recombinant IL-33 induced cutaneous and articular mechanical hypernociception in a time- and dose-dependent manner. The hypernociception was inhibited by soluble (s) ST2 (a decoy receptor of IL-33), IL-1 receptor antagonist (IL-1ra), bosentan [a dual endothelin (ET)A/ETB receptor antagonist], clazosentan (an ETA receptor antagonist), or indomethacin (a cyclooxygenase inhibitor). IL-33 induced hypernociception in IL-18−/− mice but not in TNFR1−/− or IFNγ−/− mice. The IL-33-induced hypernociception was not affected by blocking IL-15 or sympathetic amines (guanethidine). Furthermore, methylated BSA (mBSA)-induced cutaneous and articular mechanical hypernociception depended on TNFR1 and IFNγ and was blocked by sST2, IL-1ra, bosentan, clazosentan, and indomethacin. mBSA also induced significant IL-33 and ST2 mRNA expression. Importantly, we showed that mBSA induced hypernociception via the IL-33 → TNFα → IL-1β → IFNγ → ET-1 → PGE2 signaling cascade. These results therefore demonstrate that IL-33 is a key mediator of immune inflammatory hypernociception normally associated with a Th1 type of response, revealing a hitherto unrecognized function of IL-33 in a key immune pharmacological pathway that may be amenable to therapeutic intervention.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Morphine peripheral analgesia depends on activation of the PI3Kγ/AKT/nNOS/NO/KATP signaling pathway

Thiago M. Cunha; Danilo Roman-Campos; Celina M.C. Lotufo; Hugo L. Duarte; Guilherme R. Souza; Waldiceu A. Verri; Mani I. Funez; Quintino M. Dias; Ieda R. S. Schivo; Andressa C. Domingues; Daniela Sachs; Silvana Chiavegatto; Mauro M. Teixeira; John S. Hothersall; Jader Santos Cruz; Fernando Q. Cunha; Sérgio H. Ferreira

Morphine is one of the most prescribed and effective drugs used for the treatment of acute and chronic pain conditions. In addition to its central effects, morphine can also produce peripheral analgesia. However, the mechanisms underlying this peripheral action of morphine have not yet been fully elucidated. Here, we show that the peripheral antinociceptive effect of morphine is lost in neuronal nitric-oxide synthase null mice and that morphine induces the production of nitric oxide in primary nociceptive neurons. The activation of the nitric-oxide pathway by morphine was dependent on an initial stimulation of PI3Kγ/AKT protein kinase B (AKT) and culminated in increased activation of KATP channels. In the latter, this intracellular signaling pathway might cause a hyperpolarization of nociceptive neurons, and it is fundamental for the direct blockade of inflammatory pain by morphine. This understanding offers new targets for analgesic drug development.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Commensal microbiota is fundamental for the development of inflammatory pain

Flávio A. Amaral; Daniela Sachs; Vivian V. Costa; Caio T. Fagundes; Daniel Cisalpino; Thiago M. Cunha; Sérgio H. Ferreira; Fernando Q. Cunha; Tarcília Aparecida Silva; Jacques Robert Nicoli; Leda Quercia Vieira; Danielle G. Souza; Mauro M. Teixeira

The ability of an individual to sense pain is fundamental for its capacity to adapt to its environment and to avoid damage. The sensation of pain can be enhanced by acute or chronic inflammation. In the present study, we have investigated whether inflammatory pain, as measured by hypernociceptive responses, was modified in the absence of the microbiota. To this end, we evaluated mechanical nociceptive responses induced by a range of inflammatory stimuli in germ-free and conventional mice. Our experiments show that inflammatory hypernociception induced by carrageenan, lipopolysaccharide, TNF-α, IL-1β, and the chemokine CXCL1 was reduced in germ-free mice. In contrast, hypernociception induced by prostaglandins and dopamine was similar in germ-free or conventional mice. Reduction of hypernociception induced by carrageenan was associated with reduced tissue inflammation and could be reversed by reposition of the microbiota or systemic administration of lipopolysaccharide. Significantly, decreased hypernociception in germ-free mice was accompanied by enhanced IL-10 expression upon stimulation and could be reversed by treatment with an anti-IL-10 antibody. Therefore, these results show that contact with commensal microbiota is necessary for mice to develop inflammatory hypernociception. These findings implicate an important role of the interaction between the commensal microbiota and the host in favoring adaptation to environmental stresses, including those that cause pain.


Pain | 2010

IL-17 mediates articular hypernociception in antigen-induced arthritis in mice

Larissa G. Pinto; Thiago M. Cunha; Silvio M. Vieira; Henrique Lemos; Waldiceu A. Verri; Fernando Q. Cunha; Sérgio H. Ferreira

&NA; IL‐17 is an important cytokine in the physiopathology of rheumatoid arthritis (RA). However, its participation in the genesis of nociception during RA remains undetermined. In this study, we evaluated the role of IL‐17 in the genesis of articular nociception in a model of antigen (mBSA)‐induced arthritis. We found that mBSA challenge in the femur–tibial joint of immunized mice induced a dose‐ and time‐dependent mechanical hypernociception. The local IL‐17 concentration within the mBSA‐injected joints increased significantly over time. Moreover, co‐treatment of mBSA challenged mice with an antibody against IL‐17 inhibited hypernociception and neutrophil recruitment. In agreement, intraarticular injection of IL‐17 induced hypernociception and neutrophil migration, which were reduced by the pre‐treatment with fucoidin, a leukocyte adhesion inhibitor. The hypernociceptive effect of IL‐17 was also reduced in TNFR1−/− mice and by pre‐treatment with infliximab (anti‐TNF antibody), a CXCR1/2 antagonist or by an IL‐1 receptor antagonist. Consistent with these findings, we found that IL‐17 injection into joints increased the production of TNF‐&agr;, IL‐1&bgr; and CXCL1/KC. Treatment with doxycycline (non‐specific MMPs inhibitor), bosentan (ETA/ETB antagonist), indomethacin (COX inhibitor) or guanethidine (sympathetic blocker) inhibited IL‐17‐induced hypernociception. IL‐17 injection also increased PGE2 production, MMP‐9 activity and COX‐2, MMP‐9 and PPET‐1 mRNA expression in synovial membrane. These results suggest that IL‐17 is a novel pro‐nociceptive cytokine in mBSA‐induced arthritis, whose effect depends on both neutrophil migration and various pro‐inflammatory mediators, as TNF‐&agr;, IL‐1&bgr;, CXCR1/2 chemokines ligands, MMPs, endothelins, prostaglandins and sympathetic amines. Therefore, it is reasonable to propose IL‐17 targeting therapies to control this important RA symptom.


Arthritis & Rheumatism | 2012

NLRP3 inflammasome–mediated neutrophil recruitment and hypernociception depend on leukotriene B4 in a murine model of gout

Flávio A. Amaral; Vivian V. Costa; Lívia D. Tavares; Daniela Sachs; Fernanda M. Coelho; Caio T. Fagundes; Frederico M. Soriani; Tatiana N. Silveira; Larissa D. Cunha; Dario S. Zamboni; Valerie Quesniaux; Raphael S. Peres; Thiago M. Cunha; Fernando Q. Cunha; Bernhard Ryffel; Daniele G. Souza; Mauro M. Teixeira

OBJECTIVE Deposition of monosodium urate monohydrate (MSU) crystals in the joints promotes an intense inflammatory response and joint dysfunction. This study evaluated the role of the NLRP3 inflammasome and 5-lipoxygenase (5-LOX)-derived leukotriene B(4) (LTB(4) ) in driving tissue inflammation and hypernociception in a murine model of gout. METHODS Gout was induced by injecting MSU crystals into the joints of mice. Wild-type mice and mice deficient in NLRP3, ASC, caspase 1, interleukin-1β (IL-1β), IL-1 receptor type I (IL-1RI), IL-18R, myeloid differentiation factor 88 (MyD88), or 5-LOX were used. Evaluations were performed to assess neutrophil influx, LTB(4) activity, cytokine (IL-1β, CXCL1) production (by enzyme-linked immunosorbent assay), synovial microvasculature cell adhesion (by intravital microscopy), and hypernociception. Cleaved caspase 1 and production of reactive oxygen species (ROS) were analyzed in macrophages by Western blotting and fluorometric assay, respectively. RESULTS Injection of MSU crystals into the knee joints of mice induced neutrophil influx and neutrophil-dependent hypernociception. MSU crystal-induced neutrophil influx was CXCR2-dependent and relied on the induction of CXCL1 in an NLRP3/ASC/caspase 1/IL-1β/MyD88-dependent manner. LTB(4) was produced rapidly after injection of MSU crystals, and this was necessary for caspase 1-dependent IL-1β production and consequent release of CXCR2-acting chemokines in vivo. In vitro, macrophages produced LTB(4) after MSU crystal injection, and LTB(4) was relevant in the MSU crystal-induced maturation of IL-1β. Mechanistically, LTB(4) drove MSU crystal-induced production of ROS and ROS-dependent activation of the NLRP3 inflammasome. CONCLUSION These results reveal the role of the NLRP3 inflammasome in mediating MSU crystal-induced inflammation and dysfunction of the joints, and highlight a previously unrecognized role of LTB(4) in driving NLRP3 inflammasome activation in response to MSU crystals, both in vitro and in vivo.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Prostaglandin mediates IL-23/IL-17-induced neutrophil migration in inflammation by inhibiting IL-12 and IFNγ production

Henrique Lemos; Renata Grespan; Silvio M. Vieira; Thiago M. Cunha; Waldiceu A. Verri; Karla Fernandes; Fabricio O. Souto; Iain B. McInnes; Sérgio H. Ferreira; Foo Y. Liew; Fernando Q. Cunha

IL-23/IL-17-induced neutrophil recruitment plays a pivotal role in rheumatoid arthritis (RA). However, the mechanism of the neutrophil recruitment is obscure. Here we report that prostaglandin enhances the IL-23/IL-17-induced neutrophil migration in a murine model of RA by inhibiting IL-12 and IFN γ production. Methylated BSA (mBSA) and IL-23-induced neutrophil migration was inhibited by anti-IL-23 and anti-IL-17 antibodies, COX inhibitors, IL-12, or IFNγ but was enhanced by prostaglandin E2 (PGE2). IL-23-induced IL-17 production was increased by PGE2 and suppressed by COX-inhibition or IL-12. Furthermore, COX inhibition failed to reduce IL-23-induced neutrophil migration in IL-12- or IFNγ-deficient mice. IL-17-induced neutrophil migration was not affected by COX inhibitors, IL-12, or IFNγ but was inhibited by MK886 (a leukotriene synthesis inhibitor), anti-TNFα, anti-CXCL1, and anti-CXCL5 antibodies and by repertaxin (a CXCR1/2 antagonist). These treatments all inhibited mBSA- or IL-23-induced neutrophil migration. IL-17 induced neutrophil chemotaxis through a CXC chemokines-dependent pathway. Our results suggest that prostaglandin plays an important role in IL-23-induced neutrophil migration in arthritis by enhancing IL-17 synthesis and by inhibiting IL-12 and IFNγ production. We thus provide a mechanism for the pathogenic role of the IL-23/IL-17 axis in RA and also suggest an additional mechanism of action for nonsteroidal anti-inflammatory drugs.


Journal of Natural Products | 2009

Quercetin Reduces Inflammatory Pain: Inhibition of Oxidative Stress and Cytokine Production

Daniel Augusto Rodrigues Valério; Sandra R. Georgetti; Danilo A. Magro; Rubia Casagrande; Thiago M. Cunha; Fabiana T. M. C. Vicentini; Silvio M. Vieira; Maria José Vieira Fonseca; Sérgio H. Ferreira; Fernando Q. Cunha; Waldiceu A. Verri

Quercetin (1) is known to have both antioxidant and antinociceptive effects. However, the mechanism involved in its antinociceptive effect is not fully elucidated. Cytokines and reactive oxygen species have been implicated in the cascade of events resulting in inflammatory pain. Therefore, we evaluated the antinociceptive mechanism of 1 focusing on the role of cytokines and oxidative stress. Intraperitoneal and oral treatments with 1 dose-dependently inhibited inflammatory nociception induced by acetic acid and phenyl-p-benzoquinone and also the second phase of formalin- and carrageenin-induced mechanical hypernociception. Compound 1 also inhibited the hypernociception induced by cytokines (e.g., TNFalpha and CXCL1), but not by inflammatory mediators that directly sensitize the nociceptor such as PGE2 and dopamine. On the other hand, 1 reduced carrageenin-induced IL-1beta production as well as carrageenin-induced decrease of reduced glutathione (GSH) levels. These results suggest that 1 exerts its analgesic effect by inhibiting pro-nociceptive cytokine production and the oxidative imbalance mediation of inflammatory pain.


British Journal of Pharmacology | 2009

A crucial role for TNF‐α in mediating neutrophil influx induced by endogenously generated or exogenous chemokines, KC/CXCL1 and LIX/CXCL5

Silvio M. Vieira; Henrique Lemos; Renata Grespan; Marcelo Henrique Napimoga; Daniela Dal-Secco; Andressa Freitas; Thiago M. Cunha; Waldiceu A. Verri; Da Souza-Junior; Mc Jamur; Karla Fernandes; C Oliver; João Santana da Silva; Mauro M. Teixeira; Fernando Q. Cunha

Background and purpose:  Chemokines orchestrate neutrophil recruitment to inflammatory foci. In the present study, we evaluated the participation of three chemokines, KC/CXCL1, MIP‐2/CXCL2 and LIX/CXCL5, which are ligands for chemokine receptor 2 (CXCR2), in mediating neutrophil recruitment in immune inflammation induced by antigen in immunized mice.

Collaboration


Dive into the Thiago M. Cunha's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Waldiceu A. Verri

Universidade Estadual de Londrina

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rubia Casagrande

Universidade Estadual de Londrina

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ana C. Zarpelon

Universidade Estadual de Londrina

View shared research outputs
Top Co-Authors

Avatar

Jhimmy Talbot

University of São Paulo

View shared research outputs
Top Co-Authors

Avatar

Mauro M. Teixeira

Universidade Federal de Minas Gerais

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge