Network


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

Hotspot


Dive into the research topics where Fabiana S. Machado is active.

Publication


Featured researches published by Fabiana S. Machado.


Nature Medicine | 2006

Anti-inflammatory actions of lipoxin A4 and aspirin-triggered lipoxin are SOCS-2 dependent.

Fabiana S. Machado; James E Johndrow; Lisia Esper; Alexandra Dias; André Báfica; Charles N. Serhan; Julio Aliberti

Control of inflammation is crucial to prevent damage to the host during infection. Lipoxins and aspirin-triggered lipoxins are crucial modulators of proinflammatory responses; however, their intracellular mechanisms have not been completely elucidated. We previously showed that lipoxin A4 (LXA4) controls migration of dendritic cells (DCs) and production of interleukin (IL)-12 in vivo. In the absence of LXA4 biosynthetic pathways, the resulting uncontrolled inflammation during infection is lethal, despite pathogen clearance. Here we show that lipoxins activate two receptors in DCs, AhR and LXAR, and that this activation triggers expression of suppressor of cytokine signaling (SOCS)-2. SOCS-2–deficient DCs are hyper-responsive to microbial stimuli, as well as refractory to the inhibitory actions of LXA4, but not to IL-10. Upon infection with an intracellular pathogen, SOCS-2–deficient mice had uncontrolled production of proinflammatory cytokines, decreased microbial proliferation, aberrant leukocyte infiltration and elevated mortality. We also show that SOCS-2 is a crucial intracellular mediator of the anti-inflammatory actions of aspirin-induced lipoxins in vivo.


Circulation | 2000

Trypanosoma cruzi–Infected Cardiomyocytes Produce Chemokines and Cytokines That Trigger Potent Nitric Oxide–Dependent Trypanocidal Activity

Fabiana S. Machado; Gislâine A. Martins; Julio Aliberti; Fabíola Leslie Mestriner; Fernando Q. Cunha; João S. Silva

BackgroundThe pathogenesis of myocarditis that occurs in Trypanosoma cruzi–infected mice is still poorly understood. Therefore, it is important to know the mediators that trigger leukocyte migration to the heart as well as the cellular source of these possible mediators. In this study, we investigated (1) NO synthase (NOS) induction, (2) NO synthesis, (3) trypanocidal activity, and (4) chemokine and cytokine mRNA expression by isolated cardiomyocytes infected with T cruzi. Methods and ResultsMouse cardiomyocytes were isolated, infected with T cruzi, and evaluated for induction of inducible NOS (iNOS), nitrite production, trypanocidal activity, and cytokine and chemokine mRNA expression. We found that T cruzi–infected murine embryonic cardiomyocytes produced nitrite and expressed mRNAs for the chemokines chemokine growth-related oncogene, monokine induced by interferon-&ggr;, macrophage inflammatory protein-2, interferon-&ggr;–inducible protein, RANTES, and monocyte chemotactic protein, for iNOS, and for the cytokines tumor necrosis factor (TNF)-&agr; and interleukin (IL)-1&bgr;. Separate addition of IL-1&bgr;, interferon-&ggr;, TNF-&agr; or monocyte chemotactic protein, macrophage inflammatory protein-2, and interferon-&ggr;–inducible protein, to cultured cardiomyocytes resulted in NO production but low trypanocidal activity. However, simultaneous addition of IL-1&bgr;, interferon-&ggr;, and TNF-&agr; or the chemokines to cultures resulted in the induction of iNOS, high levels of nitrite, and a marked trypanocidal activity. The iNOS/l-arginine pathway mediated the latter activity, inasmuch as it was inhibited by treatment with NG-monomethyl-l-arginine. ConclusionsThese results indicate that iNOS activation and the proinflammatory cytokines and chemokines produced by cardiomyocytes are likely to control parasite growth and cell influx, thus contributing to the pathogenesis of chagasic cardiomyopathy seen in T cruzi–infected mice.


Journal of Clinical Investigation | 2005

Host control of Mycobacterium tuberculosis is regulated by 5-lipoxygenase–dependent lipoxin production

André Báfica; Charles A. Scanga; Charles N. Serhan; Fabiana S. Machado; Sandy White; Alan Sher; Julio Aliberti

Th1 type cytokine responses are critical in the control of Mycobacterium tuberculosis infection. Recent findings indicate that 5-lipoxygenase-dependent (5-LO-dependent) lipoxins regulate host IL-12 production in vivo. Here, we establish lipoxins as key chemical mediators in resistance to M. tuberculosis infection. High levels of lipoxin A4 (LXA4) were detected in sera from infected WT but not infected 5-LO-deficient mice. Moreover, lungs from M. tuberculosis-infected 5-lo-/- animals showed increased IL-12, IFN-gamma, and NO synthase 2 (NOS2) mRNA levels compared with the same tissues in WT mice. Similarly, splenocyte recall responses were enhanced in mycobacteria-infected 5-lo-/- versus WT mice. Importantly, bacterial burdens in 5-lo-/- lungs were significantly lower than those from WT mice, and this enhancement in the resistance of the 5-lo-/- animals to M. tuberculosis was completely prevented by administration of a stable LXA4 analog. Together our results demonstrate that lipoxins negatively regulate protective Th1 responses against mycobacterial infection in vivo and suggest that the inhibition of lipoxin biosynthesis could serve as a strategy for enhancing host resistance to M. tuberculosis.


Journal of Immunology | 2006

Cutting Edge: Dendritic Cells Are Essential for In Vivo IL-12 Production and Development of Resistance against Toxoplasma gondii Infection in Mice

Cheng-Hu Liu; Yu-ting Fan; Alexandra Dias; Lisia Esper; Radiah A. Corn; André Báfica; Fabiana S. Machado; Julio Aliberti

A powerful IFN-γ response is triggered upon infection with the protozoan parasite, Toxoplasma gondii. Several cell populations, including dendritic cells (DCs), macrophages, and neutrophils, produce IL-12, a key cytokine for IFN-γ induction. However, it is still unclear which of the above cell populations is its main source. Diphtheria toxin (DT) injection causes transient DC depletion in a transgenic mouse expressing Simian DT receptors under the control of the CD11c promoter, allowing us to investigate the role of DCs in IL-12 production. T. gondii-inoculated DT-treated and control groups were monitored for IL-12 levels and survival. We show in this study that DC depletion abolished IL-12 production and led to mortality. Furthermore, replenishment with wild-type, but not MyD88- or IL-12p35-deficient, DCs rescued IL-12 production, IFN-γ-induction, and resistance to infection in DC-depleted mice. Taken together, the results presented in this study indicate that DCs constitute the major IL-12-producing cell population in vivo during T. gondii infection.


Progress in Cardiovascular Diseases | 2009

Perspectives on Trypanosoma cruzi–Induced Heart Disease (Chagas Disease)

Herbert B. Tanowitz; Fabiana S. Machado; Linda A. Jelicks; Jamshid Shirani; Antonio Carlos Campos de Carvalho; David C. Spray; Stephen M. Factor; Louis V. Kirchhoff; Louis M. Weiss

Chagas disease is caused by the parasite Trypanosoma cruzi. It is a common cause of heart disease in endemic areas of Latin America. The year 2009 marks the 100th anniversary of the discovery of T cruzi infection and Chagas disease by the Brazilian physician Carlos Chagas. Chagasic cardiomyopathy develops in from 10% to 30% of persons who are chronically infected with this parasite. Echocardiography and magnetic resonance imaging (MRI) are important modalities in the evaluation and prognostication of individuals with chagasic heart disease. The etiology of chagasic heart disease likely is multifactorial. Parasite persistence, autoimmunity, and microvascular abnormalities have been studied extensively as possible pathogenic mechanisms. Experimental studies suggest that alterations in cardiac gap junctions may be etiologic in the pathogenesis of conduction abnormalities. The diagnosis of chronic Chagas disease is made by serology. The treatment of this infection has shortcomings that need to be addressed. Cardiac transplantation and bone marrow stem cell therapy for persons with Chagas disease have received increasing research attention in recent years.


The Journal of Infectious Diseases | 2005

CCR5 Plays a Critical Role in the Development of Myocarditis and Host Protection in Mice Infected with Trypanosoma cruzi

Fabiana S. Machado; Natalia S. Koyama; Vanessa Carregaro; Beatriz Rossetti Ferreira; Cristiane Maria Milanezi; Mauro M. Teixeira; Marcos A. Rossi; Joao Silva

Abstract The pathogenesis of myocarditis during Trypanosoma cruzi infection is poorly understood. We investigated the role played by chemokine receptor 5 (CCR5) in the influx of T cells to the cardiac tissue of T. cruzi—infected mice. mRNA and protein for the CCR5 ligands CCL3, CCL4, and CCL5 were detected in the hearts of infected mice in association with CD4+ and CD8+ T cells. There was a high level of CCR5 expression on CD8+ T cells in the hearts of infected mice. Moreover, CCR5 expression on CD8+ T cells was positively modulated by T. cruzi infection. CCR5-deficient mice infected with T. cruzi experienced a dramatically inhibited migration of T cells to the heart and were also more susceptible to infection. These results suggest that CCR5 and its ligands play a central role in the control of T cell influx in T. cruzi-infected mice. Knowledge of the mechanisms that trigger and control the migration of cells to the heart in patients with Chagas disease may help in the design of drugs that prevent myocarditis and protect against the development of severe disease.


Cell Cycle | 2011

Energy transfer in “parasitic” cancer metabolism: Mitochondria are the powerhouse and Achilles' heel of tumor cells

Ubaldo E. Martinez-Outschoorn; Richard G. Pestell; Anthony Howell; Mark L. Tykocinski; Fnu Nagajyothi; Fabiana S. Machado; Herbert B. Tanowitz; Federica Sotgia; Michael P. Lisanti

It is now widely recognized that the tumor microenvironment promotes cancer cell growth and metastasis via changes in cytokine secretion and extracellular matrix remodeling. However, the role of tumor stromal cells in providing energy for epithelial cancer cell growth is a newly emerging paradigm. For example, we and others have recently proposed that tumor growth and metastasis is related to an energy imbalance. Host cells produce energy-rich nutrients via catabolism (through autophagy, mitophagy, and aerobic glycolysis), which are then transferred to cancer cells to fuel anabolic tumor growth. Stromal cell-derived L-lactate is taken up by cancer cells and is used for mitochondrial oxidative phosphorylation (OXPHOS) to produce ATP efficiently. However, “parasitic” energy transfer may be a more generalized mechanism in cancer biology than previously appreciated. Two recent papers in Science and Nature Medicine now show that lipolysis in host tissues also fuels tumor growth. These studies demonstrate that free fatty acids produced by host cell lipolysis are re-used via beta-oxidation (beta-OX) in cancer cell mitochondria. Thus, stromal catabolites (such as lactate, ketones, glutamine and free fatty acids) promote tumor growth by acting as high-energy onco-metabolites. As such, host catabolism, via autophagy, mitophagy and lipolysis, may explain the pathogenesis of cancer-associated cachexia and provides exciting new druggable targets for novel therapeutic interventions. Taken together, these findings also suggest that tumor cells promote their own growth and survival by behaving as a “parasitic organism.” Hence, we propose the term “Parasitic Cancer Metabolism” to describe this type of metabolic coupling in tumors. Targeting tumor cell mitochondria (OXPHOS and beta-OX) would effectively uncouple tumor cells from their hosts, leading to their acute starvation. In this context, we discuss new evidence that high-energy onco-metabolites (produced by the stroma) can confer drug resistance. Importantly, this metabolic chemo-resistance is reversed by blocking OXPHOS in cancer cell mitochondria with drugs like Metformin, a mitochondrial “poison.” In summary, parasitic cancer metabolism is achieved architecturally by dividing tumor tissue into at least two well-defined opposing “metabolic compartments:” catabolic and anabolic.


Cellular Microbiology | 2012

Mechanisms of Trypanosoma cruzi persistence in Chagas disease

Fnu Nagajyothi; Fabiana S. Machado; Barbara A. Burleigh; Linda A. Jelicks; Philipp E. Scherer; Shankar Mukherjee; Michael P. Lisanti; Louis M. Weiss; Nisha Jain Garg; Herbert B. Tanowitz

Trypanosoma cruzi infection leads to development of chronic Chagas disease. In this article, we provide an update on the current knowledge of the mechanisms employed by the parasite to gain entry into the host cells and establish persistent infection despite activation of a potent immune response by the host. Recent studies point to a number of T. cruzi molecules that interact with host cell receptors to promote parasite invasion of the diverse host cells. T. cruzi expresses an antioxidant system and thromboxane A2 to evade phagosomal oxidative assault and suppress the hosts ability to clear parasites. Additional studies suggest that besides cardiac and smooth muscle cells that are the major target of T. cruzi infection, adipocytes and adipose tissue serve as reservoirs from where T. cruzi can recrudesce and cause disease decades later. Further, T. cruzi employs at least four strategies to maintain a symbiotic‐like relationship with the host, and ensure consistent supply of nutrients for its own survival and long‐term persistence. Ongoing and future research will continue to help refining the models of T. cruzi invasion and persistence in diverse tissues and organs in the host.


Seminars in Immunopathology | 2012

Current understanding of immunity to Trypanosoma cruzi infection and pathogenesis of Chagas disease.

Fabiana S. Machado; Walderez O. Dutra; Lisia Esper; Kenneth J. Gollob; Mauro M. Teixeira; Stephen M. Factor; Louis M. Weiss; Fnu Nagajyothi; Herbert B. Tanowitz; Nisha Jain Garg

Chagas disease caused by Trypanosoma cruzi remains an important neglected tropical disease and a cause of significant morbidity and mortality. No longer confined to endemic areas of Latin America, it is now found in non-endemic areas due to immigration. The parasite may persist in any tissue, but in recent years, there has been increased recognition of adipose tissue both as an early target of infection and a reservoir of chronic infection. The major complications of this disease are cardiomyopathy and megasyndromes involving the gastrointestinal tract. The pathogenesis of Chagas disease is complex and multifactorial involving many interactive pathways. The significance of innate immunity, including the contributions of cytokines, chemokines, reactive oxygen species, and oxidative stress, has been emphasized. The role of the components of the eicosanoid pathway such as thromboxane A2 and the lipoxins has been demonstrated to have profound effects as both pro- and anti-inflammatory factors. Additionally, we discuss the vasoconstrictive actions of thromboxane A2 and endothelin-1 in Chagas disease. Human immunity to T. cruzi infection and its role in pathogen control and disease progression have not been fully investigated. However, recently, it was demonstrated that a reduction in the anti-inflammatory cytokine IL-10 was associated with clinically significant chronic chagasic cardiomyopathy.


Frontiers in Bioscience | 2003

The role of nitric oxide in the pathogenesis of Chagas disease.

Silva Js; Fabiana S. Machado; Martins Ga

In this chapter we summarize the protective and toxic effects of nitric oxide (NO) that are frequently seen in parallel during the infection with Trypanosoma cruzi. The killing of trypomastigotes is dependent on the production of NO which is catalyzed by the inducible NO synthase (iNOS). The cytokines IFN-gamma and TNF-alpha and several chemoatractants molecules, which act on G protein-coupled serpentine receptors, are produced during the acute infection. They play major roles in the induction of iNOS, and in the NO production-dependent killing of T. cruzi by murine macrophages. On the other hand, TGF-beta and IL-10, which are also produced during the infection, are negative regulators of NO production. In addition to mediating resistance against the infection, NO can also suppress the immune response to T. cruzi via the induction of apoptosis of T cells. Furthermore, the expression of cardiac iNOS has been associated with myocardial dysfunction. In fact, we discuss here the evidences indicating that iNOS/NO pathway is involved in the pathogenesis of neuronal and myocardial dysfunction seen in patients and in experimental models.

Collaboration


Dive into the Fabiana S. Machado's collaboration.

Top Co-Authors

Avatar

Herbert B. Tanowitz

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Mauro M. Teixeira

Universidade Federal de Minas Gerais

View shared research outputs
Top Co-Authors

Avatar

Louis M. Weiss

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Fátima Brant

Universidade Federal de Minas Gerais

View shared research outputs
Top Co-Authors

Avatar

Lisia Esper

Universidade Federal de Minas Gerais

View shared research outputs
Top Co-Authors

Avatar

Julio Aliberti

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Milene Alvarenga Rachid

Universidade Federal de Minas Gerais

View shared research outputs
Top Co-Authors

Avatar

Fnu Nagajyothi

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Shankar Mukherjee

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge