Network


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

Hotspot


Dive into the research topics where André Luis Bombeiro is active.

Publication


Featured researches published by André Luis Bombeiro.


PLOS ONE | 2013

Chloroquine Treatment Enhances Regulatory T Cells and Reduces the Severity of Experimental Autoimmune Encephalomyelitis

Rodolfo Thomé; Adriel S. Moraes; André Luis Bombeiro; Alessandro S. Farias; Carolina Francelin; Thiago Alves da Costa; Rosária Di Gangi; Leonilda Maria Barbosa dos Santos; Alexandre Leite Rodrigues de Oliveira; Liana Verinaud

Background The modulation of inflammatory processes is a necessary step, mostly orchestrated by regulatory T (Treg) cells and suppressive Dendritic Cells (DCs), to prevent the development of deleterious responses and autoimmune diseases. Therapies that focused on adoptive transfer of Treg cells or their expansion in vivo achieved great success in controlling inflammation in several experimental models. Chloroquine (CQ), an anti-malarial drug, was shown to reduce inflammation, although the mechanisms are still obscure. In this context, we aimed to access whether chloroquine treatment alters the frequency of Treg cells and DCs in normal mice. In addition, the effects of the prophylactic and therapeutic treatment with CQ on Experimental Autoimmune Encephalomyelitis (EAE), an experimental model for human Multiple Sclerosis, was investigated as well. Methodology/Principal Findings EAE was induced in C57BL/6 mice by immunization with myelin oligodendrocyte glycoprotein (MOG35–55) peptide. C57BL/6 mice were intraperitoneally treated with chloroquine. Results show that the CQ treatment provoked an increase in Treg cells frequency as well as a decrease in DCs. We next evaluated whether prophylactic CQ administration is capable of reducing the clinical and histopathological signs of EAE. Our results demonstrated that CQ-treated mice developed mild EAE compared to controls that was associated with lower infiltration of inflammatory cells in the central nervous system CNS) and increased frequency of Treg cells. Also, proliferation of MOG35–55-reactive T cells was significantly inhibited by chloroquine treatment. Similar results were observed when chloroquine was administrated after disease onset. Conclusion We show for the first time that CQ treatment promotes the expansion of Treg cells, corroborating previous reports indicating that chloroquine has immunomodulatory properties. Our results also show that CQ treatment suppress the inflammation in the CNS of EAE-inflicted mice, both in prophylactic and therapeutic approaches. We hypothesized that the increased number of regulatory T cells induced by the CQ treatment is involved in the reduction of the clinical signs of EAE.


Immunology and Cell Biology | 2014

Dendritic cells treated with chloroquine modulate experimental autoimmune encephalomyelitis

Rodolfo Thomé; Luidy Kazuo Issayama; Rosaria DiGangi; André Luis Bombeiro; Thiago Alves da Costa; Isadora Tassinari Ferreira; Alexandre Leite Rodrigues de Oliveira; Liana Verinaud

Chloroquine (CQ), an antimalarial drug, has been shown to modulate the immune system and reduce the severity of experimental autoimmune encephalomyelitis (EAE). The mechanisms of disease suppression are dependent on regulatory T cell induction, although Tregs‐independent mechanisms exist. We aimed to evaluate whether CQ is capable to modulate bone marrow‐derived dendritic cells (DCs) both phenotypically and functionally as well as whether transfer of CQ‐modulated DCs reduces EAE course. Our results show that CQ‐treated DCs presented altered ultrastructure morphology and lower expression of molecules involved in antigen presentation. Consequently, T cell proliferation was diminished in coculture experiments. When transferred into EAE mice, DC‐CQ was able to reduce the clinical manifestation of the disease through the modulation of the immune response against neuroantigens. The data presented herein indicate that chloroquine‐mediated modulation of the immune system is achieved by a direct effect on DCs and that DC‐CQ adoptive transfer may be a promising approach for avoiding drug toxicity.


Brain Research Bulletin | 2014

Neuroprotective effect of tempol (4 hydroxy-tempo) on neuronal death induced by sciatic nerve transection in neonatal rats

Gabriela Bortolança Chiarotto; Luisa Drummond; Gabriela Cavarretto; André Luis Bombeiro; Alexandre Leite Rodrigues de Oliveira

Peripheral nerve injury in newborn rats triggers extensive neuronal death within the spinal cord. Because most neurodegeneration is related to oxidative stress and apoptosis, the use of antioxidants may be of therapeutic interest. Tempol is promising because of its ability to chelate reactive oxygen species and to minimize or even prevent tissue damage. Here, we evaluated neuroprotective effects of tempol following neonatal sciatic nerve transection. Two-day-old pups underwent sciatic nerve axotomy followed by tempol (12, 24 and 48 mg/kg) treatment (i.p.) at 10 min, 6 h, and every 24 h up to 1 week after injury. The rats were then killed for lumbar intumescence analysis. Nissl staining, TUNEL, synaptophysin immunolabeling and qRT-PCR (Caspase 3, Bax and Bcl2) were carried out. The results indicated that tempol treatment, at 24 mg/kg, increased up to 21% spinal cord motoneuron survival (p<0.001), also preserving pre-synaptic terminals in the neuropile. Likewise, the TUNEL-positive cell number decreased in tempol-treated animals. qRT-PCR results indicated differential increase in Caspase 3 (3-fold), Bax (13-fold) and Bcl2 (28-fold) gene expression, after 12 h following axotomy and tempol treatment. In conclusion, tempol administration has proven to be neuroprotective after neonatal nerve injury, leading to improved motoneuron survival, synapse preservation and minimizing apoptosis.


Frontiers in Cellular Neuroscience | 2016

Enhanced Immune Response in Immunodeficient Mice Improves Peripheral Nerve Regeneration Following Axotomy

André Luis Bombeiro; Júlio C. Santini; Rodolfo Thomé; Elisângela R. L. Ferreira; Sérgio Luiz Oliveira Nunes; Bárbara Monteiro Moreira; Ivan José Magayewski Bonet; Cesar Renato Sartori; Liana Verinaud; Alexandre Leite Rodrigues de Oliveira

Injuries to peripheral nerves cause loss of motor and sensory function, greatly affecting life quality. Successful repair of the lesioned nerve requires efficient cell debris removal, followed by axon regeneration and reinnervation of target organs. Such process is orchestrated by several cellular and molecular events in which glial and immune cells actively participate. It is known that tissue clearance is largely improved by macrophages, which activation is potentiated by cells and molecules of the acquired immune system, such as T helper lymphocytes and antibodies, respectively. In the present work, we evaluated the contribution of lymphocytes in the regenerative process of crushed sciatic nerves of immunocompetent (wild-type, WT) and T and B-deficient (RAG-KO) mice. In Knockout animals, we found increased amount of macrophages under basal conditions and during the initial phase of the regenerative process, that was evaluated at 2, 4, and 8 weeks after lesion (wal). That parallels with faster axonal regeneration evidenced by the quantification of neurofilament and a growth associated protein immunolabeling. The motor function, evaluated by the sciatic function index, was fully recovered in both mouse strains within 4 wal, either in a progressive fashion, as observed for RAG-KO mice, or presenting a subtle regression, as seen in WT mice between 2 and 3 wal. Interestingly, boosting the immune response by early adoptive transference of activated WT lymphocytes at 3 days after lesion improved motor recovery in WT and RAG-KO mice, which was not ameliorated when cells were transferred at 2 wal. When monitoring lymphocytes by in vivo imaging, in both mouse strains, cells migrated to the lesion site shortly after transference, remaining in the injured limb up to its complete motor recovery. Moreover, a first peak of hyperalgesia, determined by von-Frey test, was coincident with increased lymphocyte infiltration in the damaged paw. Overall, the present results suggest that a wave of immune cell infiltration takes place during subacute phase of axonal regeneration, resulting in transient set back of motor recovery following peripheral axonal injury. Moreover, modulation of the immune response can be an efficient approach to speed up nerve regeneration.


PLOS ONE | 2014

Exacerbation of Autoimmune Neuro-Inflammation in Mice Cured from Blood-Stage Plasmodium berghei Infection

Rodolfo Thomé; André Luis Bombeiro; Luidy Kazuo Issayama; Catarina Rapôso; Stefanie Costa Pinto Lopes; Thiago Alves da Costa; Rosária Di Gangi; Isadora Tassinari Ferreira; Ana Leda Longhini; Alexandre Leite Rodrigues de Oliveira; Maria Alice da Cruz Höfling; Fabio T. M. Costa; Liana Verinaud

The thymus plays an important role shaping the T cell repertoire in the periphery, partly, through the elimination of inflammatory auto-reactive cells. It has been shown that, during Plasmodium berghei infection, the thymus is rendered atrophic by the premature egress of CD4+CD8+ double-positive (DP) T cells to the periphery. To investigate whether autoimmune diseases are affected after Plasmodium berghei NK65 infection, we immunized C57BL/6 mice, which was previously infected with P.berghei NK65 and treated with chloroquine (CQ), with MOG35–55 peptide and the clinical course of Experimental Autoimmune Encephalomyelitis (EAE) was evaluated. Our results showed that NK65+CQ+EAE mice developed a more severe disease than control EAE mice. The same pattern of disease severity was observed in MOG35–55-immunized mice after adoptive transfer of P.berghei-elicited splenic DP-T cells. The higher frequency of IL-17+- and IFN-γ+-producing DP lymphocytes in the Central Nervous System of these mice suggests that immature lymphocytes contribute to disease worsening. To our knowledge, this is the first study to integrate the possible relationship between malaria and multiple sclerosis through the contribution of the thymus. Notwithstanding, further studies must be conducted to assert the relevance of malaria-induced thymic atrophy in the susceptibility and clinical course of other inflammatory autoimmune diseases.


PLOS ONE | 2016

MHC-I and PirB Upregulation in the Central and Peripheral Nervous System following Sciatic Nerve Injury.

André Luis Bombeiro; Rodolfo Thomé; Sérgio Luiz Oliveira Nunes; Bárbara Monteiro Moreira; Liana Verinaud; Alexandre Leite Rodrigues de Oliveira

Major histocompatibility complex class one (MHC-I) antigen-presenting molecules participate in central nervous system (CNS) synaptic plasticity, as does the paired immunoglobulin-like receptor B (PirB), an MHC-I ligand that can inhibit immune-cells and bind to myelin axon growth inhibitors. Based on the dual roles of both molecules in the immune and nervous systems, we evaluated their expression in the central and peripheral nervous system (PNS) following sciatic nerve injury in mice. Increased PirB and MHC-I protein and gene expression is present in the spinal cord one week after nerve transection, PirB being mostly expressed in the neuropile region. In the crushed nerve, MHC-I protein levels increased 2 weeks after lesion (wal) and progressively decreased over the next eight weeks. The same kinetics were observed for infiltrating cytotoxic T lymphocytes (CTLs) but not for PirB expression, which continuously increased. Both MHC-I and PirB were found in macrophages and Schwann cells but rarely in axons. Interestingly, at 8 wal, PirB was mainly restricted to the myelin sheath. Our findings reinforce the participation of MHC-I and PirB in CNS plasticity events. In contrast, opposing expression levels of these molecules were found in the PNS, so that MHC-I and PirB seem to be mostly implicated in antigen presentation to CTLs and axon myelination, respectively.


CNS Neuroscience & Therapeutics | 2014

Primaquine Treatment Suppresses Experimental Autoimmune Encephalomyelitis Severity

Fábio Zanucoli; Rodolfo Thomé; Amanda Pires Bonfanti; Ana Carolina de Carvalho; Luidy Kazuo Issayama; Thiago Alves da Costa; Rosária Di Gangi; Isadora Tassinari Ferreira; André Luis Bombeiro; Alexandre Leite Rodrigues de Oliveira; Liana Verinaud

Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, S~ao Paulo, BrazilCorrespondenceDr. Liana Verinaud, Department of Structuraland Functional Biology, Institute of Biology,University of Campinas (UNICAMP), RuaMonteiro Lobato, 255, Cidade Universitaria,Campinas, SP, Brazil.Tel.: +55-19-3521-6255;Fax: +55-19-3521-6276;E-mail: [email protected] 1 July 2014; revision 21 October2014; accepted 22 October 2014doi: 10.1111/cns.12357


Clinical and Vaccine Immunology | 2013

Challenge of chronically infected mice with homologous trypanosoma cruzi parasites enhances the immune response but does not modify cardiopathy: implications for the design of a therapeutic vaccine.

Christian Emerson Rosas-Jorquera; Luiz Roberto Sardinha; Fernando Delgado Pretel; André Luis Bombeiro; Maria Regina D’Império Lima; José M. Alvarez

ABSTRACT Chagas disease is a Trypanosoma cruzi-induced zoonosis that has no natural cure. Local damage induced by the parasite and the immune response causes chronic heart and digestive lesions. Efforts to develop a therapeutic vaccine that boosts the immune response to completely clear the parasite are needed because there is no effective treatment for chronically infected patients. In an attempt to modify the host-parasite equilibrium to increase parasite destruction, we analyzed cardiopathy and the immune response in chronically infected mice that were challenged with live homologous parasites. Challenge with a single dose of parasite increased CD4+ and CD8+ T cell populations, gamma interferon (IFN-γ) production, and serum-specific IgG levels. However, subpatent parasitemias and cardiac tissue were not affected. Because of the short duration of the immune boost after a single challenge, we next evaluated the impact of four parasite doses, administered 3 weeks apart. At 1 to 2 months after the last dose, the numbers of CD4+ T cells and IFN-γ-producing CD4+ memory cells and the CD4+ T cell proliferative response to T. cruzi antigen were increased in the spleen. The frequency of IFN-γ-producing CD8+ memory cells in the blood was also increased. However, the sustained challenge did not favor TH1 development; rather, it induced an increase in serum-specific IgG1 levels and mixed TH1/TH2 cytokine production. Moreover, there were no significant changes in cardiac lesions and subpatent parasitemias. In conclusion, we believe that this study may help in elucidating the necessary elements for a successful therapeutic vaccine which may reduce cardiomyopathy in chronically infected human patients.


Neuroscience Letters | 2017

Importance of major histocompatibility complex of class I (MHC-I) expression for astroglial reactivity and stability of neural circuits in vitro

André Luis Bombeiro; Rafaela Chitarra Rodrigues Hell; Gustavo Ferreira Simões; Mateus Vidigal de Castro; Alexandre Leite Rodrigues de Oliveira

MHC-I molecules are involved in the antigenic presentation of cytosol-derived peptides to CD8T lymphocytes. In the nervous system, MHC-I expression is low to absent, occurring only during certain phases of development and aging or after injuries. The involvement of MHC-I in synaptic plasticity has been reported and, following lesion, astrocytes become reactive, limiting tissue damage. Such cells also attempt to restore homeostasis by secreting cytokines and neurotrophic factors. Moreover, astrocytes modulate synapse function, by taking up and releasing neurotransmitters and by limiting the synaptic cleft. Thus, the aim of the present study was to evaluate if astrocyte activation and reactivity are related to MHC I expression and if astrogliosis can be downregulated by silencing MHC-I mRNA synthesis. Given that, we evaluated astrocyte reactivity and synaptogenesis in co-cultures of astrocytes and spinal neurons under MHC-I RNA interference. For that, the MHC-I β2-microglobulin subunit (β2m) was knocked-down by siRNA in co-cultures (β2m expression <60%, p<0.001). As measured by qRT-PCR, silencing of β2m decreased expression of the astrocytic marker GFAP (<60%, p<0.001), as well as neurotrophic factors (BDNF and GDNF) and pro-inflammatory cytokines (TNF-α, IL-1, IL-6, IL-12 and IL-17). No significant changes in synaptic stability indicate that neuron-neuron interaction was preserved after β2m silencing. Overall, the present data reinforce the importance of MHC-I expression for generation of astrogliosis, what may, in turn, become a target for future CNS/PNS therapies following injury.


European Journal of Neuroscience | 2018

Granulocyte-macrophage colony-stimulating factor improves mouse peripheral nerve regeneration following sciatic nerve crush

André Luis Bombeiro; Bruna Toledo Nunes Pereira; Alexandre Leite Rodrigues de Oliveira

Peripheral nerve injuries severely impair patients’ quality of life as full recovery is seldom achieved. Upon axonal disruption, the distal nerve stump undergoes fragmentation, and myelin breaks down; the subsequent regeneration progression is dependent on cell debris removal. In addition to tissue clearance, macrophages release angiogenic and neurotrophic factors that contribute to axon growth. Based on the importance of macrophages for nerve regeneration, especially during the initial response to injury, we treated mice with granulocyte‐macrophage colony‐stimulating factor (GM‐CSF) at various intervals after sciatic nerve crushing. Sciatic nerves were histologically analyzed at different time intervals after injury for the presence of macrophages and indicators of regeneration. Functional recovery was followed by an automated walking track test. We found that GM‐CSF potentiated early axon growth, as indicated by the enhanced expression of growth‐associated protein at 7 days postinjury. Inducible nitric oxide synthase expression increased at the beginning and at the end of the regenerative process, suggesting that nitric oxide is involved in axon growth and pruning. As expected, GM‐CSF treatment stimulated macrophage infiltration, which increased at 7 and 14 days; however, it did not improve myelin clearance. Instead, GM‐CSF stimulated early brain‐derived neurotrophic factor (BDNF) production, which peaked at 7 days. Locomotor recovery pattern was not improved by GM‐CSF treatment. The present results suggest that GM‐CSF may have beneficial effects on early axonal regeneration.

Collaboration


Dive into the André Luis Bombeiro's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Liana Verinaud

State University of Campinas

View shared research outputs
Top Co-Authors

Avatar

Rodolfo Thomé

State University of Campinas

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Luidy Kazuo Issayama

State University of Campinas

View shared research outputs
Top Co-Authors

Avatar

Rosária Di Gangi

State University of Campinas

View shared research outputs
Top Co-Authors

Avatar

Ana Leda Longhini

State University of Campinas

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge