Skip to main content
Interdisciplinary Perspectives on Infectious Diseases logoLink to Interdisciplinary Perspectives on Infectious Diseases
. 2009 Jun 14;2009:190354. doi: 10.1155/2009/190354

Oxidative Stress in Chagas Disease

Shivali Gupta 1, Jian-Jun Wen 1, Nisha Jain Garg 1, 2, 3, 4,2,3,4,*
PMCID: PMC2696642  PMID: 19547716

Abstract

There is growing evidence to suggest that chagasic myocardia are exposed to sustained oxidative stress induced injuries that may contribute to disease progression. Trypanosoma cruzi invasion- and replication-mediated cellular injuries and immune-mediated cytotoxic reactions are the common source of reactive oxygen species (ROS) during acute infection. Mitochondria are proposed to be the major source of ROS in chronic chagasic hearts. However, it has not been established yet, whether mitochondrial dysfunction is a causative factor in chagasic cardiomyopathy or a consequence of other pathological events. A better understanding of oxidative stress in relation to cardiac tissue damage would be useful in the evaluation of its true role in the pathogenesis of Chagas disease and other heart diseases. In this review, we discuss the evidence for increased oxidative stress in chagasic disease, with emphasis on mitochondrial abnormalities, and its role in sustaining oxidative stress in myocardium.

1. Chagas Disease

Chagas disease continues to pose a serious threat to health in Latin America and Mexico, and is the most important emerging parasitic disease in developed countries. According to the World Health Organization, the overall prevalence of human Trypanosoma cruzi infection is at ~16–18 million cases, and ~120 million people are at risk of infection in Latin America [1]. In most patients, the early period of T. cruzi infection goes virtually unnoticed whereas others develop an acute phase that lasts several weeks and is accompanied by such nonspecific symptoms, fever, tachycardia, weakness, and lymphadenopathy [2, 3]. After acute control of T. cruzi, infected patients enter an indeterminate phase, defined by the absence of clinical symptoms although subclinical pathology may be present. Unfortunately, 15–30 years after the initial infection, 30–40% of the infected patients develop life threatening dilated cardiomyopathy associated with clinical symptoms of ventricular dilation, arrhythmia, and cardiac arrest [4]. The pathological developments and clinical symptoms vary widely among chagasic patients [2, 57]. Not every individual infected with T. cruzi experiences the abnormalities characteristic of the three phases of Chagas disease: acute, indeterminate, and chronic. These facts make Chagas disease a complex disease and difficult to understand.

Over the years, a number of mechanisms have been proposed to explain the pathogenesis of Chagas disease (reviewed in [8, 9]). There is growing evidence to suggest that chagasic myocardia are exposed to sustained oxidative stress-induced injuries that may contribute to disease progression. In this review, we discuss the evidence for increased oxidative stress in chagasic disease, with emphasis on mitochondrial abnormalities, as well as electron transport chain dysfunction, and its role in sustaining oxidative stress in myocardium.

2. Sources of Oxidants

2.1. Overview

Broadly defined, reactive oxygen species (ROS, e.g., O2 • −, OH, and H2O2) are derivatives of molecular oxygen. ROS are unstable and react rapidly with other free radicals and macromolecules in chain reactions to generate increasingly harmful oxidants. ROS are produced through the action of specific oxidases and oxygenases (e.g., xanthine oxidase, and NADPH oxidase), peroxidases (e.g., myeloperoxidase), the Fenton reaction, and are also by-products of the electron transport chain of mitochondria [10]. Nitric oxide (NO) is produced by the enzymatic activity of nitric oxide synthases (NOS), which oxidize L-arginine, transferring electrons from NADPH. Different NOS isoforms have been identified, for example, inducible NOS (iNOS) in phagocytic cells, mtNOS in mitochondria, (eNOS) in endothelial cells, and neuronal nNOS [11].

2.2. ROS in Chagasic Hosts

During the course of T. cruzi infection and disease development, ROS can be produced as a consequence of tissue destruction caused by toxic secretions of parasite, immune-mediated cytotoxic reactions, and secondary damage to mitochondria.

In experimental studies, T. cruzi infection has been suggested to initiate ROS formation via the stimulation of inflammatory mediators, for example, cytokines and chemokines, which lead to an oxidative burst of phagocytic cells. Several investigators have used in vitro assay systems or animal models and demonstrated that T. cruzi-mediated macrophage activation results in increased levels of O2 •− formation, likely by the NADPH oxidase-dependent oxidative burst [1214]. In addition to ROS, activated macrophages can produce large amounts of NO by iNOS. Accordingly, TNF-α- and IFN-γ-dependent increased iNOS expression and NO production is noted in splenocytes of T. cruzi-infected mice [15] and in macrophages infected in vitro with T. cruzi [16]. We have found increased levels of myeloperoxidase and nitrite in the plasma of T. cruzi-infected mice [17] that are markers of neutrophil and macrophage activation, respectively. Relatively few studies have been performed to elucidate inflammatory oxidative stress in human patients. In humans, the severity of cardiac disease was correlated with high plasma levels of TNF-α and NO [18]. The NO level was also increased in indeterminate individuals in comparison to healthy controls [19]. These reactive oxidants are important for the control of T. cruzi, and may elicit toxicity to host cellular components.

Recent studies provide evidence for enhanced mitochondrial ROS generation (H2O2 and O2 •−) in chagasic myocardium. Mitochondria are the prime source of energy and many of the body's functions, including those of cardiac metabolic and contractile activities, require mitochondrial generation of ATP. Electron microscopic analysis of heart biopsies from chagasic patients and experimental animals have shown that with disease development, mitochondrial degenerative changes, that is, swelling, irregular membranes, and loss of cristae, accrue in the heart with disease development [2023]. Global microarray profiling of gene expression has identified alterations in several of the mitochondrial function related transcripts in the myocardium of infected humans [24] and experimental animals [25, 26]. The biochemical evidence for the mitochondrial dysfunction was provided by documentation of a decline in the activities of respiratory complexes, NADH-ubiquinone reductase (CI) and ubiquinol-cytochrome c reductase (CIII) [27] and ATP synthase (CV) complex [28] in chagasic murine hearts. The functional effect of these perturbations was shown by decreased mitochondrial respiration [29], and reduction in myocardial and mitochondrial ATP levels [30] in chagasic experimental models.

Imperatively, mitochondrial dysfunction also contributes to increased oxidative stress. A low, but constant, production of superoxide O2 •− occurs in mitochondria. The rate of electron leakage and O2 •− formation in mitochondria is closely related to the coupling efficiency between the respiratory chain and oxidative phosphorylation [31]. The CI and CIII complexes are the main sites for electron leakage to O2 and O2 •− generation in mitochondria [32, 33]. We have shown a decline in complex I and complex III activities in the myocardium was associated with excessive leakage of electrons to molecular oxygen and sustained ROS production in chagasic mice [27]. Further studies identified that CI was not the main source of increased ROS in chagasic hearts. Instead, defects of the myxothiazol-binding site in CIII complex resulted in enhanced electron leakage towards the Qo-center, and contributed to increased ROS generation in chagasic cardiac mitochondria [34]. Thus, conditions conducive to oxidative stress are presented in the Chagasic heart.

3. Antioxidants

3.1. Overview

The overall level of cellular ROS and its biological effects are determined by the relative rates of ROS generation and the rate of reduction by antioxidants. The principal enzymatic antioxidants are superoxide dismutase (SOD), catalase (CAT), peroxiredoxin (Prx), and glutathione peroxidase (GPx). These enzymes work in tandem to scavenge ROS. SOD exists in different isoforms, for example, manganese SOD (MnSOD) in the mitochondrial matrix and Cu- or Zn-SOD in the cytoplasm, mitochondria intermembrane space, and endothelial cell surface [35]. SOD converts O2 •− to H2O2 [36]. CAT, located in peroxisomes, converts H2O2 to H2O and O2 [37]. Prx reduces peroxides, including H2O2 and alkyl hydroperoxides [38]. The five isoforms of GPx utilize glutathione (GSH), and reduce H2O2 or lipid peroxides (ROOH) to H2O or alcohols (ROH), respectively. The byproduct of this reaction, GSSG is recycled by glutathione S reductase [38]. The nonenzymatic antioxidants, for example, vitamin E (α-tocopherol) and vitamin C (ascorbate), are abundant in aerobic organisms. Vitamin E, active in membranes, functions to reduce peroxy radicals. Vitamin C, a highly soluble antioxidant in plasma, functions by reducing α-tocopherol-lipid peroxide radicals, particularly formed in reaction with the low-density lipoproteins (LDL) [37].

3.2. Antioxidant Status in Chagasic Host

The myocardium contains high concentrations of various nonenzymatic antioxidants such as reduced glutathione (GSH) and vitamins A, C, and E, and enzymatic scavengers of ROS, including GPx and Mn- and CuZn-SOD. GSH, GPx, and MnSOD are shown to be most critical in cardiac antioxidant defenses, particularly in protecting the cardiomyocytes from oxidative injury [39, 40]. We and others have evaluated the antioxidant/oxidant balance in experimental models of chagasic disease and human patients. Our experimental studies showed that the host responds to acute T. cruzi infection by upregulating glutathione antioxidant defense constituted by GPx, GSR, and GSH. However, after the initial burst, the glutathione defense was unresponsive to chronic oxidative stress, and the cardiac levels of GSH and MnSOD were significantly diminished in chagasic mice [41]. A decline in plasma levels of GSH, the GSH/GSSG ratio [42, 43], and GPx activity [18], along with decreased MnSOD activity in PBMCs of seropositive chagasic patients [42, 43] is also noted. Decreased antioxidant levels (GPx and SOD) were correlated with an increase in TNF-α and NO levels in human patients [18]. All of these observations suggest an antioxidant response is not sufficiently activated to scavenge the ROS during progressive chagasic disease.

4. Cytotoxicity of Oxidative Stress

4.1. Overview

ROS and NO, when produced in physiological quantities, play critical roles in normal developmental processes, and control signal transduction mechanisms that regulate cell proliferation, differentiation, and death [44, 45]. However, when ROS are produced in excess or for sustained periods, they may exert toxic effects that damage cells and tissues, thereby resulting in dysfunction of physiological processes. ROS can rapidly oxidize proteins, lipids, and DNA. Lipid peroxidation causes damage to membrane integrity and loss of membrane protein function. Specifically, 4-hydroxynonenal (HNE) and malonyldialdehyde (MDA) are products of the peroxidation of membrane phospholipids [4648]. These oxidized lipids are also toxic because they are highly reactive species that result in oxidative modification of proteins [37]. For example, HNE reacts with Cys, His, or Lys residues via a Michael addition that results in irreversible alkylation and introduction of carbonyl groups into proteins [49]. The direct oxidative attack by ROS on Arg, Lys, Pro, and Thr residues can also derivatize the proteins and lead to the formation of protein carbonyls [50, 51]. NO reacts with O2 •−, to form peroxynitrite (ONOO). Myeloperoxidase-dependent oxidation of nitrite (NO2 ) results in formation of nitrogen dioxide (NO2) and nitryl chloride (NO2Cl). These reactive nitrogen species (RNS) result in protein tyrosine nitration that is widely recognized as a hallmark of nitrosative stress and inflammation [52]. Because of oxidation or nitration, a functional impairment of proteins occurs, and furthermore leads to protein turnover, for example, degradation by proteases via the proteosome [53]. DNA can be oxidized by a variety of mechanisms, resulting in nucleotide damage, for example, formation of 8-oxoguanine lesions. As a result, DNA replication may be inaccurate leading to mutations and transcription errors. While mechanisms exist to repair these DNA lesions, the level of DNA damage may exceed the capacity of the cellular repair mechanisms. Furthermore, mtDNA is believed to be particularly susceptible to sustained damage, since mitochondria may lack appropriate DNA repair mechanisms [54].

4.2. Oxidative Damage in Chagas Disease

Oxidative stress-induced injuries are a common finding in chagasic myocardium. T. cruzi has the potential to infect a wide range of host tissues [55]. As discussed above, the inflammatory infiltrate in acutely infected host is mainly constituted of phagocytic cells (e.g., macrophages) and neutrophils that produce ROS/RNS through oxidative burst [56], iNOS-dependent NO release [15], and myeloperoxidase-dependent HOCl production [57]. Oxidative damage is a consequence of the extent of oxidative stress and the antioxidant capacity. A T. cruzi-infected host does respond to inflammatory oxidative stress by an upregulation of antioxidant response constituted of GPx, GSH, and GST [41]. Yet, oxidative cellular damage, evidenced by increased protein carbonyls, MDA, and GSSG levels, is widespread, and associated with the presence of parasite foci and inflammatory infiltrate in the heart, as well as in other muscle tissues in acutely infected mice [58]. The acute oxidative damage, thus, appears to be a bystander effect of inflammatory responses elicited by T. cruzi, and occurs in all muscle tissues.

The immune control of acute parasitemia fails to provide sterile immunity. The evolution of a chronic phase is associated with mild-to-moderate diffused inflammation in different tissues and organs. It would be an oversimplification to suggest that cardiac pathology is merely an outcome of infection and inflammation, or parasite persistence that is sufficient to drive an ongoing host immune response targeted against T. cruzi. An unvarying high degree of oxidative damage persists mainly in the myocardium of chronically infected mice, as evidenced by high levels of MDA, protein carbonyl, and GSSG contents in the heart compared to findings in the skeletal muscle and colon tissue [58]. We propose the persistent activation of oxidative injurious processes plays an important role in heart-specific tissue damage in Chagas disease.

Several observations led us to consider that ROS in chronic chagasic heart are primarily produced by dysfunctional mitochondria. It is well known that ROS are generated at several subcellular sites [59] and particularly in mitochondria [60]. In effect, ~2% of the O2 consumed by mitochondria is converted to O2 •− due to spontaneous electron leaks from the respiratory chain [61]. Activated skeletal and intestinal muscles intermittently require mitochondria as an energy source, while cardiomyocytes are constantly dependent upon mitochondrial functions for their energy requirement for maintaining the contractile and other metabolic activities. According to energy demand, a ~30% cell volume of cardiomyocytes is provided by mitochondria, while in other tissues mitochondria constitute only 3–6% of cell volume [62]. Thus, maximal O2 consumption, as would be expected based upon the number of mitochondria in the heart, would produce substantial O2 •− in the heart through electron leakage from the respiratory chain. Thus, it can be inferred that even in normal conditions, heart tissue is maximally exposed to ROS of mitochondrial origin. Besides this, inefficient functioning of the respiratory complexes, as documented in chagasic hearts [27], would result in an inadequate coupling of the respiratory chain with oxidative phosphorylation and an excessive release of electrons to molecular oxygen, leading to an increased mitochondrial ROS production. We have recently found that the rate of mitochondrial O2 •− generation was substantially increased in cardiac tissue of infected mice [34], and associated with the oxidation of several subunits of the respiratory complexes [41]. The active-site thiol and heme proteins within respiratory complexes are particularly vulnerable to ROS [63]. The oxidative modification/degradation of heme proteins of the complexes release iron, the catalyst of the Fenton reaction, resulting in the formation/release of OH radicals [6466]. Taken together, these observations suggest that, under disease conditions, mitochondria are vulnerable to oxidative stress, as well as to becoming the site of an increasing order of ROS production. We, thus, propose that the acute inflammatory oxidative stress-induced mitochondrial injuries initiate a feedback cycle of ROS production and oxidative overload that causes sustained oxidative damage in the myocardium. A compromise in mitochondrial antioxidant enzyme activity (MnSOD) in chagasic myocardium would further exacerbate the mitochondrial ROS toxicity. The foregoing studies have pointed to the pathologic significance of oxidative responses in Chagasic cardiomyopathy.

It is important to note that a high degree of oxidative stress is detected in the peripheral blood of chagasic mice [58]. The demonstration of a strong positive correlation in the heart-versus-blood levels of oxidative stress markers (MDA and GSSG), and antioxidants (SOD, MnSOD, and catalase), and the mitochondrial inhibition of respiratory complexes in chronically infected mice have made it apparent that peripheral blood will be useful for understanding the role of mitochondrial decay and oxidative stress in the initiation and progression of human chagasic disease.

Subsequently, observations of increased plasma levels of GSSG and MDA and a decline in GPx activity in seropositive humans [18, 42] have led to the suggestion that chagasic patients are indeed exposed to an antioxidant/oxidant imbalance. As in experimental studies, multiple mechanisms are likely to contribute to increased oxidative stress-induced damage in chagasic patients. Plasma levels of inflammatory cytokines, NO [18] and myeloperoxidase activity [17] are increased in seropositive subjects which seems to imply that the cytotoxic effects of free radicals released by immune cells would contribute to oxidative pathology in chagasic patients. The increase in plasma MDA levels in chagasic patients may also be due to oxidatively modified lipids released as a consequence of cellular injuries, most likely, that are incurred in the cardiac tissue. This notion is supported by the observation of intense myocardial oxidative modifications [41] associated with the detection of oxidatively modified lipids and proteins in the serum [58] of mice infected by T. cruzi. Additionally, SOD and glutathione (GPx-GSH-GR) antioxidant defenses, utilized by mammalian cells to cope with free radicals [67], are found to be compromised in chagasic patients [18, 42]. These observations support the idea that glutathione antioxidant defenses, despite being active, may only be partially effective in balancing the oxidant level in chagasic patients.

5. Antioxidant Adjunct Therapy

Interventions that reduce the generation or the effects of ROS may exert beneficial effects in preventing or arresting oxidative damage. Several therapeutic interventions, for example, a vitamin E-like antioxidant, an SOD mimetic [68, 69], and an ONOO decomposition catalyst [70] have been examined for their beneficial effects against ROS in different systems. Phenyl-N-tert-butylnitrone (PBN), a nitrone-based compound, is a potent antioxidant. PBN has been shown to trap or scavenge a wide variety of free radical species, including biologically relevant O2 •− and hydroxyl OH radicals; to increase endogenous antioxidant levels; and to inhibit free radical generation [71]. In addition, PBN has been shown to inhibit the expression of a variety of inflammation-associated gene products [72].

In a recent study, we have shown that PBN treatment of infected mice prevented an oxidative stress-mediated loss in mitochondrial membrane integrity; preserved redox potential coupled with mitochondrial gene expression, and improved respiratory complex activities in infected myocardium [30]. Importantly, the PBN-mediated normalization of respiratory complex activities led to the inhibition of a feedback cycle of electron transport chain inefficiency, increased ROS production, and energy homeostasis in acute chagasic hearts [30]. Others have shown a decline in oxidative stress in human chagasic patients given Vitamin A [73]. We propose that antioxidants capable of modulating or delaying the onset of oxidative insult and mitochondrial deficiencies in the myocardium would prove to be useful in preserving cardiac functions in Chagas disease.

6. Ischemic Injury and ROS

Approximately 10% of chronic chagasic patients exhibit signs of ischemic disease [74, 75]. The abnormalities during isovolemic contraction and the early relaxation phase, in general ascribed to asynchronous onset of contraction, are noted in chagasic patients, and are similar to that seen in patients with conventional ischemic heart disease of other etiologies [76]. Others have suggested the alterations in the coronary microcirculation contribute to ischemic tissue damage in chronic chagasic patients [75, 7780]. Myocardial hypoperfusion owing to an affected microvasculature has also been noted in chagasic heart regions with normal or mildly impaired wall motion [75, 80].

Hypoxia is a critical outcome of ischemia. In hypoxic tissues, low availability of oxygen results in electron accumulation in highly reduced respiratory complexes that lead to severely compromised respiration and ATP synthesis [8183]. Ischemia also influences mitochondrial function via change in calcium flux [84], cyt c depletion (reviewed in [85]), and decline in intrinsic level of MnSOD—the mtROS scavenger [86]. The inefficient scavenging of mtROS during hypoxia is complemented by increased production of ROS at reperfusion [87]. Mitochondrial loss of cyt c is considered to potentate ROS production at reperfusion because (a) cyt c is a catalytic scavenger for mitochondrial O2 •−, and (b) loss of cyt c results in highly reduced state of respiratory complexes I, II, and III, thus, favoring electron release to molecular oxygen and O2 •− production [88, 89]. These observations suggest that mitochondrial inhibition of respiration and ATP synthesis resulting from hypoxia, coupled with an increase in O2 •− formation and ROS-induced injurious effects during reperfusion, potentially contribute to the contractile dysfunction and cell death in Chagasic hearts, to be confirmed in future studies.

7. Summary

Sustained ROS generation of inflammatory and mitochondrial origin, coupled with an inadequate antioxidant response, result in the inefficient scavenging of ROS in the heart, and lead to long-term oxidative stress, and subsequently, to oxidative damage of the cardiac cellular components during chagasic disease. The alterations in biomarkers of oxidant and antioxidant status and in respiratory complex activities in the heart and blood/plasma of infected host appear to have same pathologic tendencies, which led to the suggestion that peripheral blood would be a useful tissue for investigating the pathologic importance of impaired mitochondrial function and oxidant/antioxidant status in chagasic disease development. Further studies should examine the pathological relevance of oxidative stress in clinical severity of chronic heart disease in Chagasic patients.

Acknowledgments

The work discussed in this review was supported by Grants from the National Heart, Lung, and Blood Institute (HL088320 and HL094802) and the National Institutes of Allergy and Infectious Diseases (AI053098 and AI054578) of the National Institutes of Health, John Sealy Memorial Endowment Fund for Biomedical Research, and the American Health Assistance Foundation. The authors also would like to thank Ms. Mardelle Susman for editing the manuscript.

Abbreviations

CI:

NADH ubiquinone oxidoreductase

CII:

Succinate decylubiquinone 2, 6 dichlorophenolindophenolreductase

CIII:

Ubiquinol cytochrome c oxidoreductase

CIV:

Cytochrome c oxidase

cyt c:

Cytochrome c

GSH:

Glutathione

GPx:

Glutathione peroxidase

HNE:

4-hydroxynonenal

MDA:

Malonyldialdehyde

MPO:

Myeloperoxidase

NADH:

Nicotinamide adenine dinucleotide (reduced form)

NOS:

Nitric oxide synthase

PBN:

Phenyl-N-tert-butylnitrone

ROS:

Reactive oxygen species

SOD:

Superoxide dismutase

T. cruzi:

Trypanosoma cruzi.

References

  • 1.World Health Organization. Buenos Aires, Argentina: UNDP/World Bank/WHO; 2006. Report of the scientific working group on chagas disease. Tech. Rep. [Google Scholar]
  • 2.Brener Z. Present status of chemotherapy and chemoprophylaxis of human trypanosomiasis in the Western Hemisphere. Pharmacology & Therapeutics. 1979;7(1):71–90. doi: 10.1016/0163-7258(79)90025-1. [DOI] [PubMed] [Google Scholar]
  • 3.Milei J, Storino R. Early myocardial infarction. A feasible histologic diagnostic procedure. Japanese Heart Journal. 1986;27(3):307–319. doi: 10.1536/ihj.27.307. [DOI] [PubMed] [Google Scholar]
  • 4.Santos-Buch CA, Acosta AM. Pathology of Chagas' disease. In: Tizard I, editor. Immunology and Pathology of Trypanosomiasis. Boca Raton, Fla, USA: CRC Press; 1985. pp. 145–183. [Google Scholar]
  • 5.Andrade ZA, Andrade SG, Correa R, Sadigursky M, Ferrans VJ. Myocardial changes in acute Trypanosoma cruzi infection. Ultrastructural evidence of immune damage and the role of microangiopathy. The American Journal of Pathology. 1994;144(6):1403–1411. [PMC free article] [PubMed] [Google Scholar]
  • 6.Rassi A, Jr., Rassi A, Little WC. Chagas' heart disease. Clinical Cardiology. 2000;23(12):883–889. doi: 10.1002/clc.4960231205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.de Lourdes Higuchi M, Benvenuti LA, Reis MM, Metzger M. Pathophysiology of the heart in Chagas' disease: current status and new developments. Cardiovascular Research. 2003;60(1):96–107. doi: 10.1016/s0008-6363(03)00361-4. [DOI] [PubMed] [Google Scholar]
  • 8.Scares MBP, Pontes-De-Carvalho L, Ribeiro-Dos-Santos R. The pathogenesis of Chagas' disease: when autoimmune and parasite-specific immune responses meet. Anais da Academia Brasileira de Ciências. 2001;73(4):547–559. doi: 10.1590/s0001-37652001000400008. [DOI] [PubMed] [Google Scholar]
  • 9.Kierszenbaum F. Mechanisms of pathogenesis in Chagas disease. Acta Parasitologica. 2007;52(1):1–12. [Google Scholar]
  • 10.Turrens JF. Mitochondrial formation of reactive oxygen species. The Journal of Physiology. 2003;552(2):335–344. doi: 10.1113/jphysiol.2003.049478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Andrew PJ, Mayer B. Enzymatic function of nitric oxide synthases. Cardiovascular Research. 1999;43(3):521–531. doi: 10.1016/s0008-6363(99)00115-7. [DOI] [PubMed] [Google Scholar]
  • 12.Alvarez MN, Piacenza L, Irigoín F, Peluffo G, Radi R. Macrophage-derived peroxynitrite diffusion and toxicity to Trypanosoma cruzi . Archives of Biochemistry and Biophysics. 2004;432(2):222–232. doi: 10.1016/j.abb.2004.09.015. [DOI] [PubMed] [Google Scholar]
  • 13.Muñoz-Fernández MA, Fernández MA, Fresno M. Activation of human macrophages for the killing of intracellular Trypanosoma cruzi by TNF-α and IFN-γ through a nitric oxide-dependent mechanism. Immunology Letters. 1992;33(1):35–40. doi: 10.1016/0165-2478(92)90090-b. [DOI] [PubMed] [Google Scholar]
  • 14.Melo RC, Fabrino DL, D'Avila H, Teixeira HC, Ferreira AP. Production of hydrogen peroxide by peripheral blood monocytes and specific macrophages during experimental infection with Trypanosoma cruzi in vivo. Cell Biology International. 2003;27(10):853–861. doi: 10.1016/s1065-6995(03)00173-2. [DOI] [PubMed] [Google Scholar]
  • 15.Martins GA, Cardoso MAG, Aliberti JCS, Silva JS. Nitric oxide-induced apoptotic cell death in the acute phase of Trypanosoma cruzi infection in mice. Immunology Letters. 1998;63(2):113–120. doi: 10.1016/s0165-2478(98)00066-2. [DOI] [PubMed] [Google Scholar]
  • 16.Bergeron M, Olivier M. Trypanosoma cruzi-mediated IFN-γ-inducible nitric oxide output in macrophages is regulated by iNOS mRNA stability. The Journal of Immunology. 2006;177(9):6271–6280. doi: 10.4049/jimmunol.177.9.6271. [DOI] [PubMed] [Google Scholar]
  • 17.Dhiman M, Estrada-Franco JG, Pando JM, et al. Increased myeloperoxidase activity and protein nitration are indicators of inflammation in patients with Chagas' disease. Clinical and Vaccine Immunology. 2009;16(5):660–666. doi: 10.1128/CVI.00019-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Pérez-Fuentes R, Guégan J-F, Barnabé C, et al. Severity of chronic Chagas disease is associated with cytokine/antioxidant imbalance in chronically individuals. International Journal for Parasitology. 2003;33(3):293–299. doi: 10.1016/s0020-7519(02)00283-7. [DOI] [PubMed] [Google Scholar]
  • 19.Pérez-Fuentes R, Sánchez-Guillén MDC, González-Alvarez C, Monteón VM, Reyes PA, Rosales-Encina JL. Humoral nitric oxide levels and antibody immune response of symptomatic and indeterminate Chagas' disease patients to commercial and autochthonous Trypanosoma cruzi antigen. The American Journal of Tropical Medicine and Hygiene. 1998;58(6):715–720. doi: 10.4269/ajtmh.1998.58.715. [DOI] [PubMed] [Google Scholar]
  • 20.Carrasco Guerra HA, Palacios-Prü E, Dagert de Scorza C, Molina C, Inglessis G, Mendoza RV. Clinical, histochemical, and ultrastructural correlation in septal endomyocardial biopsies from chronic chagasic patients: detection of early myocardial damage. American Heart Journal. 1987;113(3):716–724. doi: 10.1016/0002-8703(87)90712-5. [DOI] [PubMed] [Google Scholar]
  • 21.Palacios-Pru E, Carrasco H, Scorza C, Espinoza R. Ultrastructural characteristics of different stages of human chagasic myocarditis. The American Journal of Tropical Medicine and Hygiene. 1989;41(1):29–40. [PubMed] [Google Scholar]
  • 22.Parada H, Carrasco HA, Añez N, Fuenmayor C, Inglessis I. Cardiac involvement is a constant finding in acute Chagas' disease: a clinical, parasitological and histopathological study. International Journal of Cardiology. 1997;60(1):49–54. doi: 10.1016/s0167-5273(97)02952-5. [DOI] [PubMed] [Google Scholar]
  • 23.Garg N, Popov VL, Papaconstantinou J. Profiling gene transcription reveals a deficiency of mitochondrial oxidative phosphorylation in Trypanosoma cruzi-infected murine hearts: implications in chagasic myocarditis development. Biochimica et Biophysica Acta. 2003;1638(2):106–120. doi: 10.1016/s0925-4439(03)00060-7. [DOI] [PubMed] [Google Scholar]
  • 24.Cunha-Neto E, Dzau VJ, Allen PD, et al. Cardiac gene expression profiling provides evidence for cytokinopathy as a molecular mechanism in Chagas' disease cardiomyopathy. American Journal of Pathology. 2005;167(2):305–313. doi: 10.1016/S0002-9440(10)62976-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Mukherjee S, Belbin TJ, Spray DC, et al. Microarray analysis of changes in gene expression in a murine model of chronic chagasic cardiomyopathy. Parasitology Research. 2003;91(3):187–196. doi: 10.1007/s00436-003-0937-z. [DOI] [PubMed] [Google Scholar]
  • 26.Garg N, Gerstner A, Bhatia V, DeFord J, Papaconstantinou J. Gene expression analysis in mitochondria from chagasic mice: alterations in specific metabolic pathways. Biochemical Journal. 2004;381(3):743–752. doi: 10.1042/BJ20040356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Vyatkina G, Bhatia V, Gerstner A, Papaconstantinou J, Garg N. Impaired mitochondrial respiratory chain and bioenergetics during chagasic cardiomyopathy development. Biochimica et Biophysica Acta. 2004;1689(2):162–173. doi: 10.1016/j.bbadis.2004.03.005. [DOI] [PubMed] [Google Scholar]
  • 28.Uyemura SA, Jordani MC, Polizello ACM, Curti C. Heart FoF1-ATPase changes during the acute phase of Trypanosoma cruzi infection in rats. Molecular and Cellular Biochemistry. 1996;165(2):127–133. doi: 10.1007/BF00229474. [DOI] [PubMed] [Google Scholar]
  • 29.Uyemura SA, Albuquerque S, Curti C. Energetics of heart mitochondria during acute phase of Trypanosoma cruzi infection in rats. The International Journal of Biochemistry & Cell Biology. 1995;27(11):1183–1189. doi: 10.1016/1357-2725(95)00073-x. [DOI] [PubMed] [Google Scholar]
  • 30.Wen J-J, Bhatia V, Popov VL, Garg NJ. Phenyl-α-tert-butyl nitrone reverses mitochondrial decay in acute Chagas' disease. American Journal of Pathology. 2006;169(6):1953–1964. doi: 10.2353/ajpath.2006.060475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Boveris A, Oshino N, Chance B. The cellular production of hydrogen peroxide. Biochemical Journal. 1972;128(3):617–630. doi: 10.1042/bj1280617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Ide T, Tsutsui H, Kinugawa S, et al. Mitochondrial electron transport complex I is a potential source of oxygen free radicals in the failing myocardium. Circulation Research. 1999;85(4):357–363. doi: 10.1161/01.res.85.4.357. [DOI] [PubMed] [Google Scholar]
  • 33.Chen Q, Vazquez EJ, Moghaddas S, Hoppel CL, Lesnefsky EJ. Production of reactive oxygen species by mitochondria: central role of complex III. The Journal of Biological Chemistry. 2003;278(38):36027–36031. doi: 10.1074/jbc.M304854200. [DOI] [PubMed] [Google Scholar]
  • 34.Wen JJ, Garg NJ. Mitochondrial generation of reactive oxygen species is enhanced at the Qo site of the complex III in the myocardium of Trypanosoma cruzi-infected mice: beneficial effects of an antioxidant. Journal of Bioenergetics and Biomembranes. 2008;40(6):587–598. doi: 10.1007/s10863-008-9184-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Fridovich I. Superoxide radical and superoxide dismutases. Annual Review of Biochemistry. 1995;64:97–112. doi: 10.1146/annurev.bi.64.070195.000525. [DOI] [PubMed] [Google Scholar]
  • 36.Fridovich I. Advances in Enzymology and Related Areas of Molecular Biology, Vol. 41. New York, NY, USA: John Wiley & Sons; 1974. Superoxide dismutases; pp. 35–97. [DOI] [PubMed] [Google Scholar]
  • 37.Nordberg J, Arnér ESJ. Reactive oxygen species, antioxidants, and the mammalian thioredoxin system. Free Radical Biology and Medicine. 2001;31(11):1287–1312. doi: 10.1016/s0891-5849(01)00724-9. [DOI] [PubMed] [Google Scholar]
  • 38.Chae HZ, Kang SW, Rhee SG. Isoforms of mammalian peroxiredoxin that reduce peroxides in presence of thioredoxin. Methods in Enzymology. 1999;300:219–226. doi: 10.1016/s0076-6879(99)00128-7. [DOI] [PubMed] [Google Scholar]
  • 39.Dhalla NS, Elmoselhi AB, Hata T, Makino N. Status of myocardial antioxidants in ischemia-reperfusion injury. Cardiovascular Research. 2000;47(3):446–456. doi: 10.1016/s0008-6363(00)00078-x. [DOI] [PubMed] [Google Scholar]
  • 40.Marczin N, El-Habashi N, Hoare GS, Bundy RE, Yacoub M. Antioxidants in myocardial ischemia-reperfusion injury: therapeutic potential and basic mechanisms. Archives of Biochemistry and Biophysics. 2003;420(2):222–236. doi: 10.1016/j.abb.2003.08.037. [DOI] [PubMed] [Google Scholar]
  • 41.Wen J-J, Garg N. Oxidative modification of mitochondrial respiratory complexes in response to the stress of Trypanosoma cruzi infection. Free Radical Biology and Medicine. 2004;37(12):2072–2081. doi: 10.1016/j.freeradbiomed.2004.09.011. [DOI] [PubMed] [Google Scholar]
  • 42.Wen J-J, Yachelini PC, Sembaj A, Manzur RE, Garg NJ. Increased oxidative stress is correlated with mitochondrial dysfunction in chagasic patients. Free Radical Biology and Medicine. 2006;41(2):270–276. doi: 10.1016/j.freeradbiomed.2006.04.009. [DOI] [PubMed] [Google Scholar]
  • 43.de Oliveira TB, Pedrosa RC, Filho DW. Oxidative stress in chronic cardiopathy associated with Chagas disease. International Journal of Cardiology. 2007;116(3):357–363. doi: 10.1016/j.ijcard.2006.04.046. [DOI] [PubMed] [Google Scholar]
  • 44.Finkel T. Oxidant signals and oxidative stress. Currnet Opinion in Cell Biology. 2003;15(2):247–254. doi: 10.1016/s0955-0674(03)00002-4. [DOI] [PubMed] [Google Scholar]
  • 45.Dröge W. Free radicals in the physiological control of cell function. Physiological Reviews. 2002;82(1):47–95. doi: 10.1152/physrev.00018.2001. [DOI] [PubMed] [Google Scholar]
  • 46.Valenzuela A. The biological significance of malondialdehyde determination in the assessment of tissue oxidative stress. Life Sciences. 1991;48(4):301–309. doi: 10.1016/0024-3205(91)90550-u. [DOI] [PubMed] [Google Scholar]
  • 47.Ohkawa H, Ohishi N, Yagi K. Assay for lipid peroxides in animal tissues by thiobarbituric acid reaction. Analytical Biochemistry. 1979;95(2):351–358. doi: 10.1016/0003-2697(79)90738-3. [DOI] [PubMed] [Google Scholar]
  • 48.Zarkovic N. 4-hydroxynonenal as a bioactive marker of pathophysiological processes. Molecular Aspects of Medicine. 2003;24(4-5):281–291. doi: 10.1016/s0098-2997(03)00023-2. [DOI] [PubMed] [Google Scholar]
  • 49.Uchida K, Stadtman ER. Modification of histidine residues in proteins by reaction with 4-hydroxynonenal. Proceedings of the National Academy of Sciences of the United States of America. 1992;89(10):4544–4548. doi: 10.1073/pnas.89.10.4544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Butterfield DA, Koppal T, Howard B, et al. Structural and functional changes in proteins induced by free radical-mediated oxidative stress and protective action of the antioxidants N-tert-butyl-α-phenylnitrone and vitamin E. Annals of the New York Academy of Sciences. 1998;854:448–462. doi: 10.1111/j.1749-6632.1998.tb09924.x. [DOI] [PubMed] [Google Scholar]
  • 51.Chevion M, Berenshtein E, Stadtman ER. Human studies related to protein oxidation: protein carbonyl content as a marker of damage. Free Radical Research. 2000;33(supplement):S99–S108. [PubMed] [Google Scholar]
  • 52.Schopfer FJ, Baker PRS, Freeman BA. NO-dependent protein nitration: a cell signaling event or an oxidative inflammatory response? Trends in Biochemical Sciences. 2003;28(12):646–654. doi: 10.1016/j.tibs.2003.10.006. [DOI] [PubMed] [Google Scholar]
  • 53.Floyd RA, West M, Hensley K. Oxidative biochemical markers; clues to understanding aging in long-lived species. Experimental Gerontology. 2001;36(4–6):619–640. doi: 10.1016/s0531-5565(00)00231-x. [DOI] [PubMed] [Google Scholar]
  • 54.Evans MD, Cooke MS. Factors contributing to the outcome of oxidative damage to nucleic acids. BioEssays. 2004;26(5):533–542. doi: 10.1002/bies.20027. [DOI] [PubMed] [Google Scholar]
  • 55.Younés-Chennoufi AB, Hontebeyrie-Joskowicz M, Tricottet V, Eisen H, Reynes M, Said G. Persistence of Trypanosoma cruzi antigens in the inflammatory lesions of chronically infected mice. Transactions of the Royal Society of Tropical Medicine and Hygiene. 1988;82(1):77–83. doi: 10.1016/0035-9203(88)90269-6. [DOI] [PubMed] [Google Scholar]
  • 56.Cardoni RL, Antunez MI, Morales C, Rodriguez Nantes I. Release of reactive oxygen species by phagocytic cells in response to live parasites in mice infected with Trypanosoma cruzi . The American Journal of Tropical Medicine and Hygiene. 1997;56(3):329–334. doi: 10.4269/ajtmh.1997.56.329. [DOI] [PubMed] [Google Scholar]
  • 57.Villalta F, Kierszenbaum F. Role of polymorphonuclear cells in Chagas' disease. I. Uptake and mechanisms of destruction of intracellular (amastigote) forms of Trypanosoma cruzi by human neutrophils. The Journal of Immunology. 1983;131(3):1504–1510. [PubMed] [Google Scholar]
  • 58.Wen J-J, Dhiman M, Whorton EB, Garg NJ. Tissue-specific oxidative imbalance and mitochondrial dysfunction during Trypanosoma cruzi infection in mice. Microbes and Infection. 2008;10(10-11):1201–1209. doi: 10.1016/j.micinf.2008.06.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Chance B, Sies H, Boveris A. Hydroperoxide metabolism in mammalian organs. Physiological Reviews. 1979;59(3):527–605. doi: 10.1152/physrev.1979.59.3.527. [DOI] [PubMed] [Google Scholar]
  • 60.Boveris A, Cadenas E, Stoppani AOM. Role of ubiquinone in the mitochondrial generation of hydrogen peroxide. Biochemical Journal. 1976;156(2):435–444. doi: 10.1042/bj1560435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Turrens JF. The potential of antioxidant enzymes as pharmacological agents in vivo. Xenobiotica. 1991;21(8):1033–1040. doi: 10.3109/00498259109039543. [DOI] [PubMed] [Google Scholar]
  • 62.Carvajal K, Moreno-Sánchez R. Heart metabolic disturbances in cardiovascular diseases. Archives of Medical Research. 2003;34(2):89–99. doi: 10.1016/S0188-4409(03)00004-3. [DOI] [PubMed] [Google Scholar]
  • 63.Han D, Canali R, Rettori D, Kaplowitz N. Effect of gutathione depletion on sites and topology of superoxide and hydrogen peroxide production in mitochondria. Molecular Pharmacology. 2003;64(5):1136–1144. doi: 10.1124/mol.64.5.1136. [DOI] [PubMed] [Google Scholar]
  • 64.Halliwell B, Gutteridge JMC. Oxygen toxicity, oxygen radicals, transition metals and disease. Biochemical Journal. 1984;219(1):1–14. doi: 10.1042/bj2190001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Brovko LY, Romanova NA, Ugarova NN. Bioluminescent assay of bacterial intracellular AMP, ADP, and ATP with the use of a coimmobilized three-enzyme reagent (adenylate kinase, pyruvate kinase, and firefly luciferase) Analytical Biochemistry. 1994;220(2):410–414. doi: 10.1006/abio.1994.1358. [DOI] [PubMed] [Google Scholar]
  • 66.Rush JD, Koppenol WH. Oxidizing intermediates in the reaction of ferrous EDTA with hydrogen peroxide. Reactions with organic molecules and ferrocytochrome C . The Journal of Biological Chemistry. 1986;261(15):6730–6733. [PubMed] [Google Scholar]
  • 67.Dickinson DA, Forman HJ. Glutathione in defense and signaling: lessons from a small thiol. Annals of the New York Academy of Sciences. 2002;973:488–504. doi: 10.1111/j.1749-6632.2002.tb04690.x. [DOI] [PubMed] [Google Scholar]
  • 68.Wang J, Chen H, Wang T, Diao Y, Tian K. Oxygen-derived free radicals induced cellular injury in superior mesenteric artery occlusion shock: protective effect of superoxide dismutase. Circulatory Shock. 1990;32(1):31–41. [PubMed] [Google Scholar]
  • 69.Cuzzocrea S, Costantino G, Mazzon E, De Sarro A, Caputi AP. Beneficial effects of Mn(III)tetrakis (4-benzoic acid) porphyrin (MnTBAP), a superoxide dismutase mimetic, in zymosan-induced shock. British Journal of Pharmacology. 1999;128(6):1241–1251. doi: 10.1038/sj.bjp.0702826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Salvemini D, Wang Z-Q, Zweier JL, et al. A nonpeptidyl mimic of superoxide dismutase with therapeutic activity in rats. Science. 1999;286(5438):304–306. doi: 10.1126/science.286.5438.304. [DOI] [PubMed] [Google Scholar]
  • 71.Floyd RA, Hensley K, Forster MJ, Kelleher-Anderson JA, Wood PL. Nitrones as neuroprotectants and antiaging drugs. Annals of the New York Academy of Sciences. 2002;959:321–329. doi: 10.1111/j.1749-6632.2002.tb02103.x. [DOI] [PubMed] [Google Scholar]
  • 72.Kotake Y. Pharmacologic properties of phenyl N-tert-butylnitrone. Antioxidants & Redox Signaling. 1999;1(4):481–499. doi: 10.1089/ars.1999.1.4-481. [DOI] [PubMed] [Google Scholar]
  • 73.Maçao LB, Filho DW, Pedrosa RC, et al. Antioxidant therapy attenuates oxidative stress in chronic cardiopathy associated with Chagas' disease. International Journal of Cardiology. 2007;123(1):43–49. doi: 10.1016/j.ijcard.2006.11.118. [DOI] [PubMed] [Google Scholar]
  • 74.Oliveira JSM. A natural human model of intrinsic heart nervous system denervation: Chagas' cardiopathy. American Heart Journal. 1985;110(5):1092–1098. doi: 10.1016/0002-8703(85)90222-4. [DOI] [PubMed] [Google Scholar]
  • 75.Marin-Neto JA, Simões MV, Ayres-Neto EM, et al. Studies of the coronary circulation in Chagas' heart disease. São Paulo Medical Journal. 1995;113(2):826–834. doi: 10.1590/s1516-31801995000200014. [DOI] [PubMed] [Google Scholar]
  • 76.Acquatella H, Schiller NB. Echocardiographic recognition of Chagas' disease and endomyocardial fibrosis. Journal of the American Society of Echocardiography. 1988;1(1):60–68. doi: 10.1016/s0894-7317(88)80064-6. [DOI] [PubMed] [Google Scholar]
  • 77.Rossi MA. Microvascular changes as a cause of chronic cardiomyopathy in Chagas' disease. American Heart Journal. 1990;120(1):233–236. doi: 10.1016/0002-8703(90)90191-y. [DOI] [PubMed] [Google Scholar]
  • 78.Rossi MA. Aortic endothelial cell changes in the acute septicemic phase of experimental Trypanosoma cruzi infection in rats: scanning and transmission electron microscopic study. The American Journal of Tropical Medicine and Hygiene. 1997;57(3):321–327. doi: 10.4269/ajtmh.1997.57.321. [DOI] [PubMed] [Google Scholar]
  • 79.Tanowitz HB, Kaul DK, Chen B, et al. Compromised microcirculation in acute murine Trypanosoma cruzi infection. Journal of Parasitology. 1996;82(1):124–130. [PubMed] [Google Scholar]
  • 80.Ramos SG, Rossi MA. Microcirculation and Chagas' disease: hypothesis and recent results. Revista do Instituto de Medicina Tropical de Sao Paulo. 1999;41(2):123–129. doi: 10.1590/s0036-46651999000200011. [DOI] [PubMed] [Google Scholar]
  • 81.Piper HM, Noll T, Siegmund B. Mitochondrial function in the oxygen depleted and reoxygenated myocardial cell. Cardiovascular Research. 1994;28(1):1–15. doi: 10.1093/cvr/28.1.1. [DOI] [PubMed] [Google Scholar]
  • 82.Borutaite V, Morkuniene R, Budriunaite A, et al. Kinetic analysis of changes in activity of heart mitochondrial oxidative phosphorylation system induced by ischemia. Journal of Molecular and Cellular Cardiology. 1996;28(10):2195–2201. doi: 10.1006/jmcc.1996.0211. [DOI] [PubMed] [Google Scholar]
  • 83.Jennings RB, Ganote CE. Mitochondrial structure and function in acute myocardial ischemic injury. Circulation Research. 1976;38(5) supplement 1:80–91. [PubMed] [Google Scholar]
  • 84.Ataka K, Chen D, Levitsky S, Jimenez E, Feinberg H. Effect of aging on intracellular Ca2+, pHi, and contractility during ischemia and reperfusion. Circulation. 1992;86(5) supplement 2:371–376. [PubMed] [Google Scholar]
  • 85.Borutaite V, Brown GC. Mitochondria in apoptosis of ischemic heart. FEBS Letters. 2003;541(3):1–5. doi: 10.1016/s0014-5793(03)00278-3. [DOI] [PubMed] [Google Scholar]
  • 86.Shlafer M, Myers CL, Adkins S. Mitochondrial hydrogen peroxide generation and activities of glutathione peroxidase and superoxide dismutase following global ischemia. Journal of Molecular and Cellular Cardiology. 1987;19(12):1195–1206. doi: 10.1016/s0022-2828(87)80530-8. [DOI] [PubMed] [Google Scholar]
  • 87.Lesnefsky EJ, Moghaddas S, Tandler B, Kerner J, Hoppel CL. Mitochondrial dysfunction in cardiac disease: ischemia-reperfusion, aging, and heart failure. Journal of Molecular and Cellular Cardiology. 2001;33(6):1065–1089. doi: 10.1006/jmcc.2001.1378. [DOI] [PubMed] [Google Scholar]
  • 88.Simonyan RA, Skulachev VP. Thermoregulatory uncoupling in heart muscle mitochondria: involvement of the ATP/ADP antiporter and uncoupling protein. FEBS Letters. 1998;436(1):81–84. doi: 10.1016/s0014-5793(98)01106-5. [DOI] [PubMed] [Google Scholar]
  • 89.Korshunov SS, Krasnikov BF, Pereverzev MO, Skulachev VP. The antioxidant functions of cytochrome c . FEBS Letters. 1999;462(1):192–198. doi: 10.1016/s0014-5793(99)01525-2. [DOI] [PubMed] [Google Scholar]

Articles from Interdisciplinary Perspectives on Infectious Diseases are provided here courtesy of Wiley

RESOURCES