1
|
Wang X, Chen G, Du Y, Yang J, Wang W. Transcription Factor Sox9 Exacerbates Kidney Injury through Inhibition of MicroRNA-96-5p and Activation of the Trib3/IL-6 Axis. Kidney Blood Press Res 2023; 48:611-627. [PMID: 37717559 PMCID: PMC10614512 DOI: 10.1159/000533544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 08/08/2023] [Indexed: 09/19/2023] Open
Abstract
INTRODUCTION Our study investigated the possible mechanisms of the role of the transcription factor Sox9 in the development and progression of kidney injury through regulation of the miR-96-5p/Trib3/IL-6 axis. METHODS Bioinformatics analysis was performed to identify differentially expressed genes in kidney injury and normal tissues. An in vivo animal model of kidney injury and an in vitro cellular model of kidney injury were constructed using LPS induction in 8-week-old female C57BL/6 mice and human normal renal tubular epithelial cells HK-2 for studying the possible roles of Sox9, miR-96-5p, Trib3, and IL-6 in kidney injury. RESULTS Sox9 was highly expressed in both mouse and cellular models of kidney injury. Sox9 was significantly enriched in the promoter region of miR-96-5p and repressed miR-96-5p expression. Trib3 was highly expressed in both mouse and cellular models of kidney injury and promoted inflammatory responses and kidney injury. In addition, Trib3 promoted IL-6 expression, which was highly expressed in kidney injury, and promoted the inflammatory response and extent of injury in kidney tissue. In vivo and in vitro experiments confirmed that the knockdown of Sox9 improved the inflammatory response and fibrosis of mouse kidney tissues and HK-2 cells, while the ameliorative effect of silencing Sox9 was inhibited by overexpression of IL-6. CONCLUSION Collectively, Sox9 up-regulates miR-96-5p-mediated Trib3 and activates the IL-6 signaling pathway to exacerbate the inflammatory response, ultimately promoting the development and progression of kidney injury.
Collapse
Affiliation(s)
- Xiao Wang
- Department of Urology, Fuyang People’s Hospital, Anhui Medical University, Fuyang, China
| | - Guang Chen
- Department of Urology, Fuyang People’s Hospital, Anhui Medical University, Fuyang, China
| | - Yongqiang Du
- Department of Urology, Fuyang People’s Hospital, Anhui Medical University, Fuyang, China
| | - Jiajia Yang
- Department of Urology, Fuyang People’s Hospital, Anhui Medical University, Fuyang, China
| | - Wei Wang
- Department of Urology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
2
|
Rego N, Libisch MG, Rovira C, Tosar JP, Robello C. Comparative microRNA profiling of Trypanosoma cruzi infected human cells. Front Cell Infect Microbiol 2023; 13:1187375. [PMID: 37424776 PMCID: PMC10322668 DOI: 10.3389/fcimb.2023.1187375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 06/01/2023] [Indexed: 07/11/2023] Open
Abstract
Introduction Trypanosoma cruzi, the causative agent of Chagas disease, can infect almost any nucleated cell in the mammalian host. Although previous studies have described the transcriptomic changes that occur in host cells during parasite infection, the understanding of the role of post-transcriptional regulation in this process is limited. MicroRNAs, a class of short non-coding RNAs, are key players in regulating gene expression at the post-transcriptional level, and their involvement in the host-T. cruzi interplay is a growing area of research. However, to our knowledge, there are no comparative studies on the microRNA changes that occur in different cell types in response to T. cruzi infection. Methods and results Here we investigated microRNA changes in epithelial cells, cardiomyocytes and macrophages infected with T. cruzi for 24 hours, using small RNA sequencing followed by careful bioinformatics analysis. We show that, although microRNAs are highly cell type-specific, a signature of three microRNAs -miR-146a, miR-708 and miR-1246, emerges as consistently responsive to T. cruzi infection across representative human cell types. T. cruzi lacks canonical microRNA-induced silencing mechanisms and we confirm that it does not produce any small RNA that mimics known host microRNAs. We found that macrophages show a broad response to parasite infection, while microRNA changes in epithelial and cardiomyocytes are modest. Complementary data indicated that cardiomyocyte response may be greater at early time points of infection. Conclusions Our findings emphasize the significance of considering microRNA changes at the cellular level and complement previous studies conducted at higher organizational levels, such as heart samples. While miR-146a has been previously implicated in T. cruzi infection, similarly to its involvement in many other immunological responses, miR-1246 and miR-708 are demonstrated here for the first time. Given their expression in multiple cell types, we anticipate our work as a starting point for future investigations into their role in the post-transcriptional regulation of T. cruzi infected cells and their potential as biomarkers for Chagas disease.
Collapse
Affiliation(s)
- Natalia Rego
- Unidad de Bioinformática, Institut Pasteur de Montevideo, Montevideo, Uruguay
- Laboratorio de Genómica Evolutiva, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - María Gabriela Libisch
- Laboratorio de Interacciones Hospedero Patógeno/UBM, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Carlos Rovira
- Department of Clinical Sciences Lund, Division of Oncology, Lund University, Lund, Sweden
| | - Juan Pablo Tosar
- Laboratorio de Genómica Funcional, Institut Pasteur de Montevideo, Montevideo, Uruguay
- Unidad de Bioquímica Analítica, Centro de Investigaciones Nucleares, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Carlos Robello
- Laboratorio de Interacciones Hospedero Patógeno/UBM, Institut Pasteur de Montevideo, Montevideo, Uruguay
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
3
|
Rayford KJ, Cooley A, Strode AW, Osi I, Arun A, Lima MF, Misra S, Pratap S, Nde PN. Trypanosoma cruzi dysregulates expression profile of piRNAs in primary human cardiac fibroblasts during early infection phase. Front Cell Infect Microbiol 2023; 13:1083379. [PMID: 36936778 PMCID: PMC10017870 DOI: 10.3389/fcimb.2023.1083379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 02/13/2023] [Indexed: 03/06/2023] Open
Abstract
Trypanosoma cruzi, the etiological agent of Chagas Disease, causes severe morbidity, mortality, and economic burden worldwide. Though originally endemic to Central and South America, globalization has led to increased parasite presence in most industrialized countries. About 40% of infected individuals will develop cardiovascular, neurological, and/or gastrointestinal pathologies. Accumulating evidence suggests that the parasite induces alterations in host gene expression profiles in order to facilitate infection and pathogenesis. The role of regulatory gene expression machinery during T. cruzi infection, particularly small noncoding RNAs, has yet to be elucidated. In this study, we aim to evaluate dysregulation of a class of sncRNAs called piRNAs during early phase of T. cruzi infection in primary human cardiac fibroblasts by RNA-Seq. We subsequently performed in silico analysis to predict piRNA-mRNA interactions. We validated the expression of these selected piRNAs and their targets during early parasite infection phase by stem loop qPCR and qPCR, respectively. We found about 26,496,863 clean reads (92.72%) which mapped to the human reference genome. During parasite challenge, 441 unique piRNAs were differentially expressed. Of these differentially expressed piRNAs, 29 were known and 412 were novel. In silico analysis showed several of these piRNAs were computationally predicted to target and potentially regulate expression of genes including SMAD2, EGR1, ICAM1, CX3CL1, and CXCR2, which have been implicated in parasite infection, pathogenesis, and various cardiomyopathies. Further evaluation of the function of these individual piRNAs in gene regulation and expression will enhance our understanding of early molecular mechanisms contributing to infection and pathogenesis. Our findings here suggest that piRNAs play important roles in infectious disease pathogenesis and can serve as potential biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Kayla J. Rayford
- Department of Microbiology, Immunology, and Physiology, Meharry Medical College, Nashville, TN, United States
| | - Ayorinde Cooley
- Department of Microbiology, Immunology, and Physiology, Meharry Medical College, Nashville, TN, United States
| | - Anthony W. Strode
- Department of Microbiology, Immunology, and Physiology, Meharry Medical College, Nashville, TN, United States
| | - Inmar Osi
- Department of Microbiology, Immunology, and Physiology, Meharry Medical College, Nashville, TN, United States
| | - Ashutosh Arun
- Department of Microbiology, Immunology, and Physiology, Meharry Medical College, Nashville, TN, United States
| | - Maria F. Lima
- Biomedical Sciences, School of Medicine, City College of New York, New York, NY, United States
| | - Smita Misra
- School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, United States
| | - Siddharth Pratap
- Department of Microbiology, Immunology, and Physiology, Meharry Medical College, Nashville, TN, United States
- Bioinformatics Core, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, United States
| | - Pius N. Nde
- Department of Microbiology, Immunology, and Physiology, Meharry Medical College, Nashville, TN, United States
| |
Collapse
|
4
|
Cooley A, Rayford KJ, Arun A, Villalta F, Lima MF, Pratap S, Nde PN. Trypanosoma cruzi Dysregulates piRNAs Computationally Predicted to Target IL-6 Signaling Molecules During Early Infection of Primary Human Cardiac Fibroblasts. Immune Netw 2022; 22:e51. [PMID: 36627941 PMCID: PMC9807959 DOI: 10.4110/in.2022.22.e51] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 09/20/2022] [Accepted: 10/26/2022] [Indexed: 12/31/2022] Open
Abstract
Trypanosoma cruzi, the etiological agent of Chagas disease, is an intracellular protozoan parasite, which is now present in most industrialized countries. About 40% of T. cruzi infected individuals will develop severe, incurable cardiovascular, gastrointestinal, or neurological disorders. The molecular mechanisms by which T. cruzi induces cardiopathogenesis remain to be determined. Previous studies showed that increased IL-6 expression in T. cruzi patients was associated with disease severity. IL-6 signaling was suggested to induce pro-inflammatory and pro-fibrotic responses, however, the role of this pathway during early infection remains to be elucidated. We reported that T. cruzi can dysregulate the expression of host PIWI-interacting RNAs (piRNAs) during early infection. Here, we aim to evaluate the dysregulation of IL-6 signaling and the piRNAs computationally predicted to target IL-6 molecules during early T. cruzi infection of primary human cardiac fibroblasts (PHCF). Using in silico analysis, we predict that piR_004506, piR_001356, and piR_017716 target IL6 and SOCS3 genes, respectively. We validated the piRNAs and target gene expression in T. cruzi challenged PHCF. Secreted IL-6, soluble gp-130, and sIL-6R in condition media were measured using a cytokine array and western blot analysis was used to measure pathway activation. We created a network of piRNAs, target genes, and genes within one degree of biological interaction. Our analysis revealed an inverse relationship between piRNA expression and the target transcripts during early infection, denoting the IL-6 pathway targeting piRNAs can be developed as potential therapeutics to mitigate T. cruzi cardiomyopathies.
Collapse
Affiliation(s)
- Ayorinde Cooley
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN 37208, USA
| | - Kayla J. Rayford
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN 37208, USA
| | - Ashutosh Arun
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN 37208, USA
| | - Fernando Villalta
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN 37208, USA
- Department of Cell, Molecular, and Biomedical Sciences, School of Medicine, The City College of New York, New York, NY 10031, USA
| | - Maria F. Lima
- Department of Cell, Molecular, and Biomedical Sciences, School of Medicine, The City College of New York, New York, NY 10031, USA
| | - Siddharth Pratap
- School of Graduate Studies and Research, Meharry Medical College, Nashville, TN 37208, USA
| | - Pius N. Nde
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN 37208, USA
| |
Collapse
|
5
|
Zhou Q, Meng D, Li F, Zhang X, Liu L, Zhu Y, Liu S, Xu M, Deng J, Lei Z, Sluijter JP, Xiao J. Inhibition of HIPK2 protects stress-induced pathological cardiac remodeling. EBioMedicine 2022; 85:104274. [PMID: 36182775 PMCID: PMC9526139 DOI: 10.1016/j.ebiom.2022.104274] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 09/05/2022] [Accepted: 09/05/2022] [Indexed: 11/26/2022] Open
|
6
|
Xanthine Analogs Suppress Trypanosoma cruzi Infection In Vitro Using PDEs as Targets. MICROBIOLOGY RESEARCH 2022. [DOI: 10.3390/microbiolres13040052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Trypanosoma cruzi (T. cruzi), the causative agent of Chagas disease, has infected 6 million people, putting 70 million people at risk worldwide. Presently, very limited drugs are available, and these have severe side effects. Hence, there is an urgency to delve into other pathways and targets for novel drugs. Trypanosoma cruzi (T. cruzi) expresses a number of different cyclic AMP (cAMP)-specific phosphodiesterases (PDEs). cAMP is one of the key regulators of mammalian cell proliferation and differentiation, and it also plays an important role in T. cruzi growth. Very few studies have demonstrated the important role of cyclic nucleotide-specific PDEs in T. cruzi’s survival. T. cruzi phosphodiesterase C (TcrPDEC) has been proposed as a potential new drug target for treating Chagas disease. In the current study, we screen several analogs of xanthine for potency against trypomastigote and amastigote growth in vitro using three different strains of T. cruzi (Tulahuen, Y and CA-1/CL72). One of the potent analogs, GVK14, has been shown to inhibit all three strains of amastigotes in host cells as well as axenic cultures. In conclusion, xanthine analogs that inhibit T. cruzi PDE may provide novel alternative therapeutic options for Chagas disease.
Collapse
|
7
|
Oliveira TGM, Venturini G, Alvim JM, Feijó LL, Dinardo CL, Sabino EC, Seidman JG, Seidman CE, Krieger JE, Pereira AC. Different Transcriptomic Response to T. cruzi Infection in hiPSC-Derived Cardiomyocytes From Chagas Disease Patients With and Without Chronic Cardiomyopathy. Front Cell Infect Microbiol 2022; 12:904747. [PMID: 35873155 PMCID: PMC9301326 DOI: 10.3389/fcimb.2022.904747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 06/16/2022] [Indexed: 11/16/2022] Open
Abstract
Chagas disease is a tropical zoonosis caused by Trypanosoma cruzi. After infection, the host present an acute phase, usually asymptomatic, in which an extensive parasite proliferation and intense innate immune activity occurs, followed by a chronic phase, characterized by low parasitemia and development of specific immunity. Most individuals in the chronic phase remain without symptoms or organ damage, a state called indeterminate IND form. However, 20 to 40% of individuals develop cardiac or gastrointestinal complications at any time in life. Cardiomyocytes have an important role in the development of Chronic Chagas Cardiomyopathy (CCC) due to transcriptional and metabolic alterations that are crucial for the parasite survival and replication. However, it still not clear why some infected individuals progress to a cardiomyopathy phase, while others remain asymptomatic. In this work, we used hiPSCs-derived cardiomyocytes (hiPSC-CM) to investigate patterns of infection, proliferation and transcriptional response in IND and CCC patients. Our data show that T. cruzi infection and proliferation efficiency do not differ significantly in PBMCs and hiPSC-CM from both groups. However, RNA-seq analysis in hiPSC-CM infected for 24 hours showed a significantly different transcriptional response to the parasite in cells from IND or CCC patients. Cardiomyocytes from IND showed significant differences in the expression of genes related to antigen processing and presentation, as well as, immune co-stimulatory molecules. Furthermore, the downregulation of collagen production genes and extracellular matrix components was significantly different in these cells. Cardiomyocytes from CCC, in turn, showed increased expression of mTORC1 pathway and unfolded protein response genes, both associated to increased intracellular ROS production. These data point to a differential pattern of response, determined by baseline genetic differences between groups, which may have an impact on the development of a chronic outcome with or without the presentation of cardiac symptoms.
Collapse
Affiliation(s)
- Theo G. M. Oliveira
- Laboratório de Genética e Cardiologia Molecular, Instituto do Coração (InCor), Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HC/FMUSP), São Paulo, Brazil
- Instituto do Coração (InCor), Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HC/FMUSP), São Paulo, Brazil
- Fundação Pró-Sangue Hemocentro de São Paulo, Divisão de Pesquisa – São Paulo, SP, Brazil
- *Correspondence: Theo G. M. Oliveira,
| | - Gabriela Venturini
- Laboratório de Genética e Cardiologia Molecular, Instituto do Coração (InCor), Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HC/FMUSP), São Paulo, Brazil
- Genetics Department, Harvard Medical School, MA, United States
| | - Juliana M. Alvim
- Laboratório de Genética e Cardiologia Molecular, Instituto do Coração (InCor), Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HC/FMUSP), São Paulo, Brazil
| | - Larissa L. Feijó
- Fundação Pró-Sangue Hemocentro de São Paulo, Divisão de Pesquisa – São Paulo, SP, Brazil
| | - Carla L. Dinardo
- Fundação Pró-Sangue Hemocentro de São Paulo, Divisão de Pesquisa – São Paulo, SP, Brazil
| | - Ester C. Sabino
- Instituto do Medicina Tropical (IMT), Universidade de São Paulo – São Paulo, SP, Brazil
| | | | - Christine E. Seidman
- Genetics Department, Harvard Medical School, MA, United States
- Cardiovascular Division, Brigham and Women’s Hospital, & Harvard Medical School, Boston, MA, United States
- Howard Hughes Medical Institute, Chevy Chase, MD, United States
| | - Jose E. Krieger
- Laboratório de Genética e Cardiologia Molecular, Instituto do Coração (InCor), Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HC/FMUSP), São Paulo, Brazil
- Instituto do Coração (InCor), Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HC/FMUSP), São Paulo, Brazil
| | - Alexandre C. Pereira
- Laboratório de Genética e Cardiologia Molecular, Instituto do Coração (InCor), Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HC/FMUSP), São Paulo, Brazil
- Instituto do Coração (InCor), Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HC/FMUSP), São Paulo, Brazil
- Genetics Department, Harvard Medical School, MA, United States
| |
Collapse
|
8
|
Jones KM, Poveda C, Versteeg L, Bottazzi ME, Hotez PJ. Preclinical advances and the immunophysiology of a new therapeutic chagas disease vaccine. Expert Rev Vaccines 2022; 21:1185-1203. [PMID: 35735065 DOI: 10.1080/14760584.2022.2093721] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Chronic infection with the protozoal parasite Trypanosoma cruzi leads to a progressive cardiac disease, known as chronic Chagasic cardiomyopathy (CCC). A new therapeutic Chagas disease vaccine is in development to augment existing antiparasitic chemotherapy drugs. AREAS COVERED We report on our current understanding of the underlying immunologic and physiologic mechanisms that lead to CCC, including parasite immune escape mechanisms that allow persistence and the subsequent inflammatory and fibrotic processes that lead to clinical disease. We report on vaccine design and the observed immunotherapeutic effects including induction of a balanced TH1/TH2/TH17 immune response that leads to reduced parasite burdens and tissue pathology. Further, we report vaccine-linked chemotherapy, a dose sparing strategy to further reduce parasite burdens and tissue pathology. EXPERT OPINION Our vaccine-linked chemotherapeutic approach is a multimodal treatment strategy, addressing both the parasite persistence and the underlying deleterious host inflammatory and fibrotic responses that lead to cardiac dysfunction. In targeting treatment towards patients with chronic indeterminate or early determinate Chagas disease, this vaccine-linked chemotherapeutic approach will be highly economical and will reduce the global disease burden and deaths due to CCC.
Collapse
Affiliation(s)
- Kathryn M Jones
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America.,Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Cristina Poveda
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America.,Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Leroy Versteeg
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America.,Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America.,Cell Biology and Immunology Group, Wageningen University & Research, De Elst 1, 6708 WD Wageningen, The Netherlands
| | - Maria Elena Bottazzi
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America.,Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America.,Department of Biology, Baylor University, Waco, Texas, United States of America
| | - Peter J Hotez
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America.,Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America.,Department of Biology, Baylor University, Waco, Texas, United States of America.,James A. Baker III Institute for Public Policy, Rice University, Houston, Texas, United States of America.,Hagler Institute for Advanced Study at Texas A&M University, College Station, Texas, United States of America
| |
Collapse
|
9
|
Arun A, Rayford KJ, Cooley A, Rana T, Rachakonda G, Villalta F, Pratap S, Lima MF, Sheibani N, Nde PN. Thrombospondin-1 expression and modulation of Wnt and hippo signaling pathways during the early phase of Trypanosoma cruzi infection of heart endothelial cells. PLoS Negl Trop Dis 2022; 16:e0010074. [PMID: 34986160 PMCID: PMC8730400 DOI: 10.1371/journal.pntd.0010074] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 12/08/2021] [Indexed: 12/13/2022] Open
Abstract
The protozoan parasite, Trypanosoma cruzi, causes severe morbidity and mortality in afflicted individuals. Approximately 30% of T. cruzi infected individuals present with cardiac pathology. The invasive forms of the parasite are carried in the vascular system to infect other cells of the body. During transportation, the molecular mechanisms by which the parasite signals and interact with host endothelial cells (EC) especially heart endothelium is currently unknown. The parasite increases host thrombospondin-1 (TSP1) expression and activates the Wnt/β-catenin and hippo signaling pathways during the early phase of infection. The links between TSP1 and activation of the signaling pathways and their impact on parasite infectivity during the early phase of infection remain unknown. To elucidate the significance of TSP1 function in YAP/β-catenin colocalization and how they impact parasite infectivity during the early phase of infection, we challenged mouse heart endothelial cells (MHEC) from wild type (WT) and TSP1 knockout mice with T. cruzi and evaluated Wnt signaling, YAP/β-catenin crosstalk, and how they affect parasite infection. We found that in the absence of TSP1, the parasite induced the expression of Wnt-5a to a maximum at 2 h (1.73±0.13), P< 0.001 and enhanced the level of phosphorylated glycogen synthase kinase 3β at the same time point (2.99±0.24), P<0.001. In WT MHEC, the levels of Wnt-5a were toned down and the level of p-GSK-3β was lowest at 2 h (0.47±0.06), P< 0.01 compared to uninfected control. This was accompanied by a continuous significant increase in the nuclear colocalization of β-catenin/YAP in TSP1 KO MHEC with a maximum Pearson correlation coefficient of (0.67±0.02), P< 0.05 at 6 h. In WT MHEC, the nuclear colocalization of β-catenin/YAP remained steady and showed a reduction at 6 h (0.29±0.007), P< 0.05. These results indicate that TSP1 plays an important role in regulating β-catenin/YAP colocalization during the early phase of T. cruzi infection. Importantly, dysregulation of this crosstalk by pre-incubation of WT MHEC with a β-catenin inhibitor, endo-IWR 1, dramatically reduced the level of infection of WT MHEC. Parasite infectivity of inhibitor treated WT MHEC was similar to the level of infection of TSP1 KO MHEC. These results indicate that the β-catenin pathway induced by the parasite and regulated by TSP1 during the early phase of T. cruzi infection is an important potential therapeutic target, which can be explored for the prophylactic prevention of T. cruzi infection.
Collapse
Affiliation(s)
- Ashutosh Arun
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Kayla J. Rayford
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Ayorinde Cooley
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Tanu Rana
- Department of Professional Medical Education and Molecular Biology Core Facility, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Girish Rachakonda
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Fernando Villalta
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Siddharth Pratap
- School of Graduate Studies and Research, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Maria F. Lima
- School of Graduate Studies and Research, Meharry Medical College, Nashville, Tennessee, United States of America
- Department of Molecular and Cellular and Biomedical Sciences, School of Medicine, The City College of New York, New York, United States of America
| | - Nader Sheibani
- Department of Ophthalmology and Visual Sciences, Biomedical Engineering, and Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Pius N. Nde
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, Tennessee, United States of America
| |
Collapse
|
10
|
Benzo(a)pyrene-induced cytotoxicity, cell proliferation, DNA damage, and altered gene expression profiles in HT-29 human colon cancer cells. Cell Biol Toxicol 2021; 37:891-913. [PMID: 33411230 DOI: 10.1007/s10565-020-09579-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 12/25/2020] [Indexed: 12/18/2022]
Abstract
In the US alone, around 60,000 lives/year are lost to colon cancer. In order to study the mechanisms of colon carcinogenesis, in vitro model systems are required in addition to in vivo models. Towards this end, we have used the HT-29 colon cancer cells, cultured in Dulbecco's Modified Eagle Medium (DMEM), which were exposed to benzo(a)pyrene (BaP), a ubiquitous and prototypical environmental and dietary toxicant at 1, 10, 100 nM and 1, 5, 10, and 25 μM concentrations for 96 h. Post-BaP exposure, growth, cytotoxicity, apoptosis, and cell cycle changes were determined. The BaP metabolite concentrations in colon cells were identified and measured. Furthermore, the BaP biotransformation enzymes were studied at the protein and mRNA levels. The BaP exposure-induced damage to DNA was assessed by measuring the oxidative damage to DNA and the concentrations of BaP-DNA adducts. To determine the whole repertoire of genes that are up- or downregulated by BaP exposure, mRNA transcriptome analysis was conducted. There was a BaP exposure concentration (dose)-dependent decrease in cell growth, cytotoxicity, and modulation of the cell cycle in the treatment groups compared to untreated or dimethylsulfoxide (DMSO: vehicle for BaP)-treated categories. The phase I biotransformation enzymes, CYP1A1 and 1B1, showed BaP concentration-dependent expression. On the other hand, phase II enzymes did not exhibit any marked variation. Consistent with the expression of phase I enzymes, elevated concentrations of BaP metabolites were generated, contributing to the formation of DNA lesions and stable DNA adducts, which were also BaP concentration-dependent. In summary, our studies established that biotransformation of BaP contributes to cytotoxicity, proliferation of tumor cells, and alteration of gene expression by BaP. • Benzo(a)pyrene (BaP) is an environmental and dietary toxicant. • BaP causes cytotoxicity in cultured HT-29 colon cancer cells. • mRNA transcriptome analyses revealed that BaP impacts cell growth, cell cycle, biotransformation, and DNA damage.
Collapse
|
11
|
Rodríguez-Angulo HO, Colombet-Naranjo D, Maza MC, Poveda C, Herreros-Cabello A, Mendoza I, Perera JC, Goyo JD, Gironès N, Fresno M. Molecular Remodeling of Cardiac Sinus Node Associated with Acute Chagas Disease Myocarditis. Microorganisms 2021; 9:microorganisms9112208. [PMID: 34835334 PMCID: PMC8620628 DOI: 10.3390/microorganisms9112208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/13/2021] [Accepted: 10/19/2021] [Indexed: 01/04/2023] Open
Abstract
Chagas disease principally affects Latin-American people, but it currently has worldwide distribution due to migration. Death among those with Chagas disease can occur suddenly and without warning, even in those who may not have evidence of clinical or structural cardiac disease and who are younger than 60 years old. HCN4 channels, one of the principal elements responsible for pacemaker currents, are associated with cardiac fetal reprogramming and supraventricular and ventricular arrhythmias, but their role in chagasic arrhythmias is not clear. We found that a single-dose administration of ivabradine, which blocks HCN4, caused QTc and QRS enlargement and an increase in P-wave amplitude and was associated with ventricular and supraventricular arrhythmias in mice challenged with isoproterenol, a chronotropic/ionotropic positive agent. Continuous treatment with ivabradine did not alter the QTc interval, but P-wave morphology was deeply modified, generating supraventricular arrhythmias. In addition, we found that repolarization parameters improved with ivabradine treatment. These effects could have been caused by the high HCN4 expression observed in auricular and ventricular tissue in infected mice. Thus, we suggest, for the first time, that molecular remodeling by overexpression of HCN4 channels may be related to supraventricular arrhythmias in acute Chagas disease, causing ivabradine over-response. Thus, ivabradine treatment should be administered with caution, while HCN4 overexpression may be an indicator of heart failure and/or sudden death risk.
Collapse
Affiliation(s)
- Héctor O. Rodríguez-Angulo
- Instituto Venezolano de Investigaciones Científicas, Caracas 1020A, Venezuela; (H.O.R.A.); (D.C.-N.)
- Unidad de Biología Celular, Departamento de Ciencias Morfológicas, Programa de Medicina, Facultad de Ciencias de la vida, Universidad Centroccidental Lisandro Alvarado, Barquisimeto 3001, Venezuela; (J.C.P.); (J.D.G.)
| | - Diana Colombet-Naranjo
- Instituto Venezolano de Investigaciones Científicas, Caracas 1020A, Venezuela; (H.O.R.A.); (D.C.-N.)
| | - María C. Maza
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, 28049 Madrid, Spain; (M.C.M.); (C.P.); (A.-H.C.)
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, 28049 Madrid, Spain
| | - Cristina Poveda
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, 28049 Madrid, Spain; (M.C.M.); (C.P.); (A.-H.C.)
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, 28049 Madrid, Spain
| | - Alfonso Herreros-Cabello
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, 28049 Madrid, Spain; (M.C.M.); (C.P.); (A.-H.C.)
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, 28049 Madrid, Spain
| | - Iván Mendoza
- Instituto de Medicina Tropical, Universidad Central de Venezuela, Caracas 1060, Venezuela;
| | - Juan C. Perera
- Unidad de Biología Celular, Departamento de Ciencias Morfológicas, Programa de Medicina, Facultad de Ciencias de la vida, Universidad Centroccidental Lisandro Alvarado, Barquisimeto 3001, Venezuela; (J.C.P.); (J.D.G.)
| | - Juan D. Goyo
- Unidad de Biología Celular, Departamento de Ciencias Morfológicas, Programa de Medicina, Facultad de Ciencias de la vida, Universidad Centroccidental Lisandro Alvarado, Barquisimeto 3001, Venezuela; (J.C.P.); (J.D.G.)
| | - Núria Gironès
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, 28049 Madrid, Spain; (M.C.M.); (C.P.); (A.-H.C.)
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, 28049 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital Universitario de la Princesa, 28009 Madrid, Spain
- Correspondence: (N.G.); (M.F.)
| | - Manuel Fresno
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, 28049 Madrid, Spain; (M.C.M.); (C.P.); (A.-H.C.)
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, 28049 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital Universitario de la Princesa, 28009 Madrid, Spain
- Correspondence: (N.G.); (M.F.)
| |
Collapse
|
12
|
Hoffman KA, Villar MJ, Poveda C, Bottazzi ME, Hotez PJ, Tweardy DJ, Jones KM. Signal Transducer and Activator of Transcription-3 Modulation of Cardiac Pathology in Chronic Chagasic Cardiomyopathy. Front Cell Infect Microbiol 2021; 11:708325. [PMID: 34504808 PMCID: PMC8421853 DOI: 10.3389/fcimb.2021.708325] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 07/23/2021] [Indexed: 01/31/2023] Open
Abstract
Chronic Chagasic cardiomyopathy (CCC) is a severe clinical manifestation that develops in 30%–40% of individuals chronically infected with the protozoal parasite Trypanosoma cruzi and is thus an important public health problem. Parasite persistence during chronic infection drives pathologic changes in the heart, including myocardial inflammation and progressive fibrosis, that contribute to clinical disease. Clinical manifestations of CCC span a range of symptoms, including cardiac arrhythmias, thromboembolic disease, dilated cardiomyopathy, and heart failure. This study aimed to investigate the role of signal transducer and activator of transcription-3 (STAT3) in cardiac pathology in a mouse model of CCC. STAT3 is a known cellular mediator of collagen deposition and fibrosis. Mice were infected with T. cruzi and then treated daily from 70 to 91 days post infection (DPI) with TTI-101, a small molecule inhibitor of STAT3; benznidazole; a combination of benznidazole and TTI-101; or vehicle alone. Cardiac function was evaluated at the beginning and end of treatment by echocardiography. By the end of treatment, STAT3 inhibition with TTI-101 eliminated cardiac fibrosis and fibrosis biomarkers but increased cardiac inflammation; serum levels of interleukin-6 (IL-6), and IFN−γ; cardiac gene expression of STAT1 and nuclear factor-κB (NF-κB); and upregulation of IL-6 and Type I and Type II IFN responses. Concurrently, decreased heart function was measured by echocardiography and myocardial strain. These results indicate that STAT3 plays a critical role in the cardiac inflammatory–fibrotic axis during CCC.
Collapse
Affiliation(s)
- Kristyn A Hoffman
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States.,Department of Pediatrics, Section of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Maria Jose Villar
- Department of Pediatrics, Section of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States.,Texas Children's Hospital Center for Vaccine Development, Houston, TX, United States
| | - Cristina Poveda
- Department of Pediatrics, Section of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States.,Texas Children's Hospital Center for Vaccine Development, Houston, TX, United States
| | - Maria Elena Bottazzi
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States.,Department of Pediatrics, Section of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States.,Texas Children's Hospital Center for Vaccine Development, Houston, TX, United States.,Department of Biology, Baylor University, Waco, TX, United States
| | - Peter J Hotez
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States.,Department of Pediatrics, Section of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States.,Texas Children's Hospital Center for Vaccine Development, Houston, TX, United States.,Department of Biology, Baylor University, Waco, TX, United States
| | - David J Tweardy
- Department of Infectious Diseases, Infection Control & Employee Health, Division of Internal Medicine and Department of Molecular & Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Kathryn M Jones
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States.,Department of Pediatrics, Section of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States.,Texas Children's Hospital Center for Vaccine Development, Houston, TX, United States
| |
Collapse
|
13
|
Cruz-Saavedra L, Vallejo GA, Guhl F, Messenger LA, Ramírez JD. Transcriptional remodeling during metacyclogenesis in Trypanosoma cruzi I. Virulence 2021; 11:969-980. [PMID: 32715914 PMCID: PMC7549971 DOI: 10.1080/21505594.2020.1797274] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Metacyclogenesis is one of the most important processes in the life cycle of Trypanosoma cruzi. In this stage, noninfective epimastigotes become infective metacyclic trypomastigotes. However, the transcriptomic changes that occur during this transformation remain uncertain. Illumina RNA-sequencing of epimastigotes and metacyclic trypomastigotes belonging to T. cruzi DTU I was undertaken. Sequencing reads were aligned and mapped against the reference genome, differentially expressed genes between the two life cycle stages were identified, and metabolic pathways were reconstructed. Gene expression differed significantly between epimastigotes and metacyclic trypomastigotes. The cellular pathways that were mostly downregulated during metacyclogenesis involved glucose energy metabolism (glycolysis, pyruvate metabolism, the Krebs cycle, and oxidative phosphorylation), amino acid metabolism, and DNA replication. By contrast, the processes where an increase in gene expression was observed included those related to autophagy (particularly Atg7 and Atg8 transcripts), corroborating its importance during metacyclogenesis, endocytosis, by an increase in the expression of the AP-2 complex subunit alpha, protein processing in the endoplasmic reticulum and meiosis. Study findings indicate that in T. cruzi metacyclic trypomastigotes, metabolic processes are decreased, and expression of genes involved in specific cell cycle processes is increased to facilitate transformation to this infective stage.
Collapse
Affiliation(s)
- Lissa Cruz-Saavedra
- Grupo de Investigaciones Microbiológicas-UR (GIMUR), Departamento de Biología, Facultad de Ciencias Naturales, Universidad del Rosario , Bogotá, Colombia
| | - Gustavo A Vallejo
- Laboratorio de Investigaciones en Parasitología Tropical, Facultad de Ciencias, Universidad del Tolima , Ibagué, Colombia
| | - Felipe Guhl
- Centro de Investigaciones en Microbiología y Parasitología Tropical (CIMPAT), Facultad de Ciencias, Universidad de Los Andes , Bogotá, Colombia
| | | | - Juan David Ramírez
- Grupo de Investigaciones Microbiológicas-UR (GIMUR), Departamento de Biología, Facultad de Ciencias Naturales, Universidad del Rosario , Bogotá, Colombia
| |
Collapse
|
14
|
Guo HH, Jing XY, Chen H, Xu HX, Zhu BM. STAT3 but Not STAT5 Contributes to the Protective Effect of Electroacupuncture Against Myocardial Ischemia/Reperfusion Injury in Mice. Front Med (Lausanne) 2021; 8:649654. [PMID: 34307396 PMCID: PMC8299366 DOI: 10.3389/fmed.2021.649654] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 05/13/2021] [Indexed: 02/05/2023] Open
Abstract
Electroacupuncture (EA) can help reduce infarct size and injury resulting from myocardial ischemia/reperfusion (I/R); however, the underlying molecular mechanism remains unknown. We previously reported that STAT5 plays a critical role in the cardioprotective effect of remote ischemic preconditioning (RIPC). Here, we assessed the effects of electroacupuncture pretreatment (EAP) on myocardial I/R injury in the presence and/or absence of Stat5 in mice and investigated whether EAP exerts its cardioprotective effects in a STAT5-dependent manner. Adult Stat5fl/fl and Stat5-cKO mice were exposed to EAP at Neiguan (PC6) for 7 days before the induction of I/R injury by left anterior descending (LAD) coronary artery ligation. The myocardial infarct size (IS), area at risk, and apoptotic rate of cardiomyocytes were detected. RT-qPCR and western blotting were used to measure gene and protein expression, respectively, in homogenized heart tissues. RNA-seq was used to identify candidate genes and pathways. Our results showed that EAP decreased IS and the rate of cardiomyocyte apoptosis. We further found that STAT5 was activated by EAP in Stat5fl/fl mice but not in Stat5-cKO mice, whereas the opposite was observed for STAT3. Following EAP, the levels of the antiapoptotic proteins Bcl-xL, Bcl-2, and p-AKT were increased in the presence of Stat5, while that of interleukin 10 (IL-10) was increased in both Stat5fl/fl and Stat5-cKO. The gene expression profile in heart tissues was different between Stat5fl/fl and the Stat5-cKO mice with EAP. Importantly, the top 30 DEGs under EAP in the Stat5-cKO mice were enriched in the IL-6/STAT3 signaling pathway. Our results revealed for the first time that the protective effect of EAP following myocardial I/R injury was attributable to, but not dependent on, STAT5. Additionally, we found that EAP could activate STAT3 signaling in the absence of the Stat5 gene, and could also activate antiapoptotic, survival, and anti-inflammatory signaling pathways.
Collapse
Affiliation(s)
- Hui-Hui Guo
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xin-Yue Jing
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hui Chen
- Rehabilitation Medicine Department, YE DA Hospital of Yantai, Yantai, China
| | - Hou-Xi Xu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Bing-Mei Zhu
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
15
|
Hoffman K, Liu Z, Hossain E, Bottazzi ME, Hotez PJ, Jones KM, McCall LI. Alterations to the Cardiac Metabolome Induced by Chronic T. cruzi Infection Relate to the Degree of Cardiac Pathology. ACS Infect Dis 2021; 7:1638-1649. [PMID: 33843195 PMCID: PMC8588157 DOI: 10.1021/acsinfecdis.0c00816] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Chronic Chagasic cardiomyopathy (CCC) is a Neglected Tropical Disease caused by the parasite Trypanosoma cruzi. The pathognomonic findings in symptomatic CCC patients and animal models includes diffuse cardiac fibrosis and inflammation with persistent parasite presence in the heart. This study investigated chemical alterations in different regions of the heart in relation to cardiac pathology indicators to better understand the long-term pathogenesis of this neglected disease. We used data from echocardiography, fibrosis biomarkers, and histopathological analysis to fully evaluate cardiac pathology. Metabolites isolated from the pericardial and endocardial sides of the right ventricular myocardium were analyzed by liquid chromatography tandem mass spectrometry. The endocardial sections contained significantly less cardiac inflammation and fibrosis than the pericardial sections. Cardiac levels of acylcarnitines, phosphocholines, and other metabolites were significantly disrupted in accordance with cardiac fibrosis, inflammation, and serum fibrosis biomarker levels. These findings have potential implications in treatment and monitoring for CCC patients.
Collapse
Affiliation(s)
- Kristyn Hoffman
- Department of Molecular Virology and Microbiology and Department of Pediatrics, Section of Tropical Medicine, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Zongyuan Liu
- Department of Chemistry and Biochemistry and Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Ekram Hossain
- Department of Chemistry and Biochemistry and Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Maria Elena Bottazzi
- Department of Molecular Virology and Microbiology and Department of Pediatrics, Section of Tropical Medicine, Baylor College of Medicine, Houston, Texas 77030, United States; Texas Children’s Hospital Center for Vaccine Development, Houston, Texas 77030, United States; Department of Biology, Baylor University, Waco, Texas 76798, United States
| | - Peter J. Hotez
- Department of Molecular Virology and Microbiology and Department of Pediatrics, Section of Tropical Medicine, Baylor College of Medicine, Houston, Texas 77030, United States; Texas Children’s Hospital Center for Vaccine Development, Houston, Texas 77030, United States; Department of Biology, Baylor University, Waco, Texas 76798, United States
| | - Kathryn M. Jones
- Department of Molecular Virology and Microbiology and Department of Pediatrics, Section of Tropical Medicine, Baylor College of Medicine, Houston, Texas 77030, United States; Texas Children’s Hospital Center for Vaccine Development, Houston, Texas 77030, United States
| | - Laura-Isobel McCall
- Department of Chemistry and Biochemistry, Laboratories of Molecular Anthropology and Microbiome Research, and Department of Microbiology and Plant Biology, University of Oklahoma, Norman, Oklahoma 73019, United States
| |
Collapse
|
16
|
piRNAs as Modulators of Disease Pathogenesis. Int J Mol Sci 2021; 22:ijms22052373. [PMID: 33673453 PMCID: PMC7956838 DOI: 10.3390/ijms22052373] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/24/2021] [Accepted: 02/24/2021] [Indexed: 12/20/2022] Open
Abstract
Advances in understanding disease pathogenesis correlates to modifications in gene expression within different tissues and organ systems. In depth knowledge about the dysregulation of gene expression profiles is fundamental to fully uncover mechanisms in disease development and changes in host homeostasis. The body of knowledge surrounding mammalian regulatory elements, specifically regulators of chromatin structure, transcriptional and translational activation, has considerably surged within the past decade. A set of key regulators whose function still needs to be fully elucidated are small non-coding RNAs (sncRNAs). Due to their broad range of unfolding functions in the regulation of gene expression during transcription and translation, sncRNAs are becoming vital to many cellular processes. Within the past decade, a novel class of sncRNAs called PIWI-interacting RNAs (piRNAs) have been implicated in various diseases, and understanding their complete function is of vital importance. Historically, piRNAs have been shown to be indispensable in germline integrity and stem cell development. Accumulating research evidence continue to reveal the many arms of piRNA function. Although piRNA function and biogenesis has been extensively studied in Drosophila, it is thought that they play similar roles in vertebrate species, including humans. Compounding evidence suggests that piRNAs encompass a wider functional range than small interfering RNAs (siRNAs) and microRNAs (miRNAs), which have been studied more in terms of cellular homeostasis and disease. This review aims to summarize contemporary knowledge regarding biogenesis, and homeostatic function of piRNAs and their emerging roles in the development of pathologies related to cardiomyopathies, cancer, and infectious diseases.
Collapse
|
17
|
Libisch MG, Rego N, Robello C. Transcriptional Studies on Trypanosoma cruzi - Host Cell Interactions: A Complex Puzzle of Variables. Front Cell Infect Microbiol 2021; 11:692134. [PMID: 34222052 PMCID: PMC8248493 DOI: 10.3389/fcimb.2021.692134] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 05/26/2021] [Indexed: 01/05/2023] Open
Abstract
Chagas Disease, caused by the protozoan parasite Trypanosoma cruzi, affects nearly eight million people in the world. T. cruzi is a complex taxon represented by different strains with particular characteristics, and it has the ability to infect and interact with almost any nucleated cell. The T. cruzi-host cell interactions will trigger molecular signaling cascades in the host cell that will depend on the particular cell type and T. cruzi strain, and also on many different experimental variables. In this review we collect data from multiple transcriptomic and functional studies performed in different infection models, in order to highlight key differences between works that in our opinion should be addressed when comparing and discussing results. In particular, we focus on changes in the respiratory chain and oxidative phosphorylation of host cells in response to infection, which depends on the experimental model of T. cruzi infection. Finally, we also discuss host cell responses which reiterate independently of the strain, cell type and experimental conditions.
Collapse
Affiliation(s)
- María Gabriela Libisch
- Laboratorio de Interacciones Hospedero Patógeno-UBM, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Natalia Rego
- Unidad de Bioinformática, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Carlos Robello
- Laboratorio de Interacciones Hospedero Patógeno-UBM, Institut Pasteur de Montevideo, Montevideo, Uruguay
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
- *Correspondence: Carlos Robello,
| |
Collapse
|
18
|
Trypanosoma cruzi Modulates PIWI-Interacting RNA Expression in Primary Human Cardiac Myocytes during the Early Phase of Infection. Int J Mol Sci 2020; 21:ijms21249439. [PMID: 33322418 PMCID: PMC7764157 DOI: 10.3390/ijms21249439] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/28/2020] [Accepted: 12/10/2020] [Indexed: 02/07/2023] Open
Abstract
Trypanosoma cruzi dysregulates the gene expression profile of primary human cardiomyocytes (PHCM) during the early phase of infection through a mechanism which remains to be elucidated. The role that small non-coding RNAs (sncRNA) including PIWI-interacting RNA (piRNA) play in regulating gene expression during the early phase of infection is unknown. To understand how T. cruzi dysregulate gene expression in the heart, we challenged PHCM with T. cruzi trypomastigotes and analyzed sncRNA, especially piRNA, by RNA-sequencing. The parasite induced significant differential expression of host piRNAs, which can target and regulate the genes which are important during the early infection phase. An average of 21,595,866 (88.40%) of clean reads mapped to the human reference genome. The parasite induced 217 unique piRNAs that were significantly differentially expressed (q ≥ 0.8). Of these differentially expressed piRNAs, 6 were known and 211 were novel piRNAs. In silico analysis showed that some of the dysregulated known and novel piRNAs could target and potentially regulate the expression of genes including NFATC2, FOS and TGF-β1, reported to play important roles during T. cruzi infection. Further evaluation of the specific functions of the piRNAs in the regulation of gene expression during the early phase of infection will enhance our understanding of the molecular mechanism of T. cruzi pathogenesis. Our novel findings constitute the first report that T. cruzi can induce differential expression of piRNAs in PHCM, advancing our knowledge about the involvement of piRNAs in an infectious disease model, which can be exploited for biomarker and therapeutic development.
Collapse
|
19
|
Strauss M, Casares-Marfil D, Alcaraz A, Palma-Vega M, Bosch-Nicolau P, Lo Presti MS, Molina I, González CI, Martín J, Acosta-Herrera M. Lack of Association of IL6 polymorphism with the susceptibility to Chagas disease in Latin American populations. Acta Trop 2020; 210:105546. [PMID: 32492396 DOI: 10.1016/j.actatropica.2020.105546] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 05/04/2020] [Accepted: 05/16/2020] [Indexed: 11/26/2022]
Abstract
The aim of the present study was to analyze IL6 rs1800795 genetic variant in the susceptibility to Trypanosoma cruzi infection and in the development of chronic Chagas cardiomyopathy (CCC), in five independent Latin American cohorts. A total of 3,087 individuals from Latin American countries (Argentina, Bolivia, Peru, and two cohorts from Colombia) were studied. In all cohorts, patients were classified as seropositive for T. cruzi antigens (n= 1,963) and seronegative (n= 1,124). Based on clinical evaluation, the seropositive patients, were classified as CCC (n= 900) and asymptomatic (n= 1,063). No statistically significant differences in the frequency of IL6 rs1800795 between seropositive and seronegative, or between CCC and asymptomatic patients, were found. Furthermore, after the meta-analysis no statistically significant differences were observed. Our results do not support a contribution of IL6 rs1800795 genetic variant in the susceptibility to the infection and the development of chronic Chagas cardiomyopathy in the studied populations.
Collapse
Affiliation(s)
- Mariana Strauss
- Centro de Estudios e Investigación de la Enfermedad de Chagas y Leishmaniasis, FCM, INICSA-CONICET-UNC, Córdoba, Argentina.
| | - Desiré Casares-Marfil
- Instituto de Parasitología y Biomedicina López-Neyra, IPBLN-CSIC, PTS Granada, Granada, España.
| | - Alexia Alcaraz
- Instituto de Parasitología y Biomedicina López-Neyra, IPBLN-CSIC, PTS Granada, Granada, España
| | - Miriam Palma-Vega
- Instituto de Parasitología y Biomedicina López-Neyra, IPBLN-CSIC, PTS Granada, Granada, España
| | - Pau Bosch-Nicolau
- Unidad de Medicina Tropical y Salud Internacional Hospital UniversitariValld'Hebron, PROSICS, Barcelona, España
| | - María Silvina Lo Presti
- Centro de Estudios e Investigación de la Enfermedad de Chagas y Leishmaniasis, FCM, INICSA-CONICET-UNC, Córdoba, Argentina
| | - Israel Molina
- Unidad de Medicina Tropical y Salud Internacional Hospital UniversitariValld'Hebron, PROSICS, Barcelona, España
| | | | - Javier Martín
- Instituto de Parasitología y Biomedicina López-Neyra, IPBLN-CSIC, PTS Granada, Granada, España
| | | | | |
Collapse
|
20
|
Oliveira AER, Pereira MCA, Belew AT, Ferreira LRP, Pereira LMN, Neves EGA, Nunes MDCP, Burleigh BA, Dutra WO, El-Sayed NM, Gazzinelli RT, Teixeira SMR. Gene expression network analyses during infection with virulent and avirulent Trypanosoma cruzi strains unveil a role for fibroblasts in neutrophil recruitment and activation. PLoS Pathog 2020; 16:e1008781. [PMID: 32810179 PMCID: PMC7508367 DOI: 10.1371/journal.ppat.1008781] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 09/22/2020] [Accepted: 07/07/2020] [Indexed: 12/24/2022] Open
Abstract
Chagas disease is caused by Trypanosoma cruzi, a protozoan parasite that has a heterogeneous population composed of a pool of strains with distinct characteristics, including variable levels of virulence. In previous work, transcriptome analyses of parasite genes after infection of human foreskin fibroblasts (HFF) with virulent (CL Brener) and non-virulent (CL-14) clones derived from the CL strain, revealed a reduced expression of genes encoding parasite surface proteins in CL-14 compared to CL Brener during the final steps of the intracellular differentiation from amastigotes to trypomastigotes. Here we analyzed changes in the expression of host genes during in vitro infection of HFF cells with the CL Brener and CL-14 strains by analyzing total RNA extracted from cells at 60 and 96 hours post-infection (hpi) with each strain, as well as from uninfected cells. Similar transcriptome profiles were observed at 60 hpi with both strains compared to uninfected samples. However, at 96 hpi, significant differences in the number and expression levels of several genes, particularly those involved with immune response and cytoskeleton organization, were observed. Further analyses confirmed the difference in the chemokine/cytokine signaling involved with the recruitment and activation of immune cells such as neutrophils upon T. cruzi infection. These findings suggest that infection with the virulent CL Brener strain induces a more robust inflammatory response when compared with the non-virulent CL-14 strain. Importantly, the RNA-Seq data also exposed an unexplored role of fibroblasts as sentinel cells that may act by recruiting neutrophils to the initial site of infection. This role for fibroblasts in the regulation of the inflammatory response during infection by T. cruzi was corroborated by measurements of levels of different chemokines/cytokines during in vitro infection and in plasma from Chagas disease patients as well as by neutrophil activation and migration assays. Trypanosoma cruzi is the causative agent of Chagas disease, a debilitating and often life-threatening illness that affects 6 to 7 million people mainly in Latin America. The parasite, transmitted to humans by an insect vector, needs to invade different cells from the infected person in order to multiply and spread the infection to various organs, including the heart and the gut. In this study, we investigated how the host cell responds to the infection by analyzing changes in the expression of human genes in fibroblasts infected with the CL Brener and CL-14 strains, which are strains that present highly distinct virulent phenotypes during infection in mice. We showed that human fibroblasts build a strong immune response upon infection by T. cruzi and that this response is different depending on the parasite strain: infection with the virulent CL Brener strain induces a more robust inflammatory response compared with the infection with the avirulent CL-14 strain. We also showed that, in response to the infection, fibroblasts produce molecules that can recruit and activate neutrophils, which are important immune cells that controls the infection.
Collapse
Affiliation(s)
- Antonio Edson R. Oliveira
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Milton C. A. Pereira
- Centro de Pesquisas Rene Rachou, Fundação Oswaldo Cruz, Belo Horizonte, MG, Brazil
| | - Ashton T. Belew
- Department of Cell Biology and Molecular Genetics and Center for Bioinformatics and Computational Biology, University of Maryland, College Park, Maryland, United States of America
| | - Ludmila R. P. Ferreira
- Departamento de Morfologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Eula G. A. Neves
- Departamento de Morfologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Maria do Carmo P. Nunes
- Departamento de Clínica Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Barbara A. Burleigh
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Walderez O. Dutra
- Departamento de Morfologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Najib M. El-Sayed
- Department of Cell Biology and Molecular Genetics and Center for Bioinformatics and Computational Biology, University of Maryland, College Park, Maryland, United States of America
| | - Ricardo T. Gazzinelli
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
- Centro de Pesquisas Rene Rachou, Fundação Oswaldo Cruz, Belo Horizonte, MG, Brazil
- * E-mail: (SMRT); (RTG)
| | - Santuza M. R. Teixeira
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
- * E-mail: (SMRT); (RTG)
| |
Collapse
|
21
|
Arun A, Rayford KJ, Cooley A, Rachakonda G, Villalta F, Pratap S, Lima MF, Sheibani N, Nde PN. Thrombospondin-1 Plays an Essential Role in Yes-Associated Protein Nuclear Translocation during the Early Phase of Trypanosoma cruzi Infection in Heart Endothelial Cells. Int J Mol Sci 2020; 21:ijms21144912. [PMID: 32664627 PMCID: PMC7403984 DOI: 10.3390/ijms21144912] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/09/2020] [Accepted: 07/09/2020] [Indexed: 01/03/2023] Open
Abstract
The protozoan parasite Trypanosoma cruzi is the causative agent of Chagas disease. This neglected tropical disease causes severe morbidity and mortality in endemic regions. About 30% of T. cruzi infected individuals will present with cardiac complications. Invasive trypomastigotes released from infected cells can be carried in the vascular endothelial system to infect neighboring and distant cells. During the process of cellular infection, the parasite induces host cells, to increase the levels of host thrombospondin-1 (TSP-1), to facilitate the process of infection. TSP-1 plays important roles in the functioning of vascular cells, including vascular endothelial cells with important implications in cardiovascular health. Many signal transduction pathways, including the yes-associated protein 1 (YAP)/transcriptional coactivator, with PDZ-binding motif (TAZ) signaling, which are upstream of TSP-1, have been linked to the pathophysiology of heart damage. The molecular mechanisms by which T. cruzi signals, and eventually infects, heart endothelial cells remain unknown. To evaluate the importance of TSP-1 expression in heart endothelial cells during the process of T. cruzi infection, we exposed heart endothelial cells prepared from Wild Type and TSP-1 Knockout mouse to invasive T. cruzi trypomastigotes at multiple time points, and evaluated changes in the hippo signaling cascade using immunoblotting and immunofluorescence assays. We found that the parasite turned off the hippo signaling pathway in TSP-1KO heart endothelial cells. The levels of SAV1 and MOB1A increased to a maximum of 2.70 ± 0.23 and 5.74 ± 1.45-fold at 3 and 6 h, respectively, in TSP-1KO mouse heart endothelial cells (MHEC), compared to WT MHEC, following a parasite challenge. This was accompanied by a significant continuous increase in the nuclear translocation of downstream effector molecule YAP, to a maximum mean nuclear fluorescence intensity of 10.14 ± 0.40 at 6 h, compared to wild type cells. Furthermore, we found that increased nuclear translocated YAP significantly colocalized with the transcription co-activator molecule pan-TEAD, with a maximum Pearson's correlation coefficient of 0.51 ± 0.06 at 6 h, compared to YAP-Pan-TEAD colocalization in the WT MHEC, which decreased significantly, with a minimum Pearson's correlation coefficient of 0.30 ± 0.01 at 6 h. Our data indicate that, during the early phase of infection, upregulated TSP-1 is essential for the regulation of the hippo signaling pathway. These studies advance our understanding of the molecular interactions occurring between heart endothelial cells and T. cruzi, in the presence and absence of TSP-1, providing insights into processes linked to parasite dissemination and pathogenesis.
Collapse
Affiliation(s)
- Ashutosh Arun
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN 37208, USA; (A.A.); (K.J.R.); (A.C.); (G.R.); (F.V.)
| | - Kayla J. Rayford
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN 37208, USA; (A.A.); (K.J.R.); (A.C.); (G.R.); (F.V.)
| | - Ayorinde Cooley
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN 37208, USA; (A.A.); (K.J.R.); (A.C.); (G.R.); (F.V.)
| | - Girish Rachakonda
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN 37208, USA; (A.A.); (K.J.R.); (A.C.); (G.R.); (F.V.)
| | - Fernando Villalta
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN 37208, USA; (A.A.); (K.J.R.); (A.C.); (G.R.); (F.V.)
| | - Siddharth Pratap
- School of Graduate Studies and Research, Meharry Medical College, Nashville, TN 37208, USA; (S.P.); (M.F.L.)
| | - Maria F. Lima
- School of Graduate Studies and Research, Meharry Medical College, Nashville, TN 37208, USA; (S.P.); (M.F.L.)
- Department of Molecular Cellular and Biomedical Sciences, School of Medicine, The City College of New York, New York, NY 10031, USA
| | - Nader Sheibani
- Department of Ophthalmology and Visual Sciences, Biomedical Engineering and Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA;
| | - Pius N. Nde
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN 37208, USA; (A.A.); (K.J.R.); (A.C.); (G.R.); (F.V.)
- Correspondence: ; Tel.: +1-615-327-6997
| |
Collapse
|
22
|
Villanueva-Lizama LE, Cruz-Chan JV, Versteeg L, Teh-Poot CF, Hoffman K, Kendricks A, Keegan B, Pollet J, Gusovsky F, Hotez PJ, Bottazzi ME, Jones KM. TLR4 agonist protects against Trypanosoma cruzi acute lethal infection by decreasing cardiac parasite burdens. Parasite Immunol 2020; 42:e12769. [PMID: 32592180 DOI: 10.1111/pim.12769] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/18/2020] [Accepted: 06/19/2020] [Indexed: 01/11/2023]
Abstract
E6020 is a synthetic agonist of Toll-like receptor-4 (TLR4). The purpose of this study was to evaluate the effect of different doses of E6020-SE on Trypanosoma cruzi-specific immune responses and its ability to confer protection against acute lethal infection in mice. Forty female BALB/c were infected with 500 trypomastigotes of T cruzi H1 strain, divided into four groups (n = 10) and treated at 7- and 14-day post-infection (dpi) with different doses of E6020-SE or PBS (control). Survival was followed for 51 days, mice were euthanized and hearts were collected to evaluate parasite burden, inflammation and fibrosis. We found significantly higher survival and lower parasite burdens in mice injected with E6020-SE at all doses compared to the control group. However, E6020-SE treatment did not significantly reduce cardiac inflammation or fibrosis. On the other hand, E6020-SE modulated Th1 and Th2 cytokines, decreasing IFN-γ and IL-4 in a dose-dependent manner after stimulation with parasite antigens. We conclude that E6020-SE alone increased survival by decreasing cardiac parasite burdens in BALB/c mice acutely infected with T cruzi but failed to prevent cardiac damage. Our results suggest that for optimal protection, a vaccine antigen is necessary to balance and orient a protective immune response.
Collapse
Affiliation(s)
- Liliana E Villanueva-Lizama
- Department of Pediatrics and National School of Tropical Medicine, Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA.,Laboratorio de Parasitología, Centro de Investigaciones Regionales Dr. Hideyo Noguchi, Universidad Autónoma de Yucatán, Mérida, México
| | - Julio V Cruz-Chan
- Department of Pediatrics and National School of Tropical Medicine, Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA.,Laboratorio de Parasitología, Centro de Investigaciones Regionales Dr. Hideyo Noguchi, Universidad Autónoma de Yucatán, Mérida, México
| | - Leroy Versteeg
- Department of Pediatrics and National School of Tropical Medicine, Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA.,Cell Biology and Immunology Group, Wageningen University & Research, Wageningen, The Netherlands
| | - Christian F Teh-Poot
- Laboratorio de Parasitología, Centro de Investigaciones Regionales Dr. Hideyo Noguchi, Universidad Autónoma de Yucatán, Mérida, México
| | - Kristyn Hoffman
- Department of Pediatrics and National School of Tropical Medicine, Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA
| | - April Kendricks
- Department of Pediatrics and National School of Tropical Medicine, Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA
| | - Brian Keegan
- Department of Pediatrics and National School of Tropical Medicine, Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA
| | - Jeroen Pollet
- Department of Pediatrics and National School of Tropical Medicine, Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA
| | | | - Peter J Hotez
- Department of Pediatrics and National School of Tropical Medicine, Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA.,Department of Biology, Baylor University, Waco, TX, USA.,James A. Baker III Institute for Public Policy, Rice University, Houston, TX, USA.,Hagler Institute for Advanced Study at Texas A&M University, College Station, TX, USA
| | - Maria Elena Bottazzi
- Department of Pediatrics and National School of Tropical Medicine, Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA.,Department of Biology, Baylor University, Waco, TX, USA
| | - Kathryn M Jones
- Department of Pediatrics and National School of Tropical Medicine, Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
23
|
Wozniak JM, Silva TA, Thomas D, Siqueira-Neto JL, McKerrow JH, Gonzalez DJ, Calvet CM. Molecular dissection of Chagas induced cardiomyopathy reveals central disease associated and druggable signaling pathways. PLoS Negl Trop Dis 2020; 14:e0007980. [PMID: 32433643 PMCID: PMC7279607 DOI: 10.1371/journal.pntd.0007980] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 06/08/2020] [Accepted: 03/25/2020] [Indexed: 12/13/2022] Open
Abstract
Chagas disease, the clinical presentation of T. cruzi infection, is a major human health concern. While the acute phase of Chagas disease is typically asymptomatic and self-resolving, chronically infected individuals suffer numerous sequelae later in life. Cardiomyopathies in particular are the most severe consequence of chronic Chagas disease and cannot be reversed solely by parasite load reduction. To prioritize new therapeutic targets, we unbiasedly interrogated the host signaling events in heart tissues isolated from a Chagas disease mouse model using quantitative, multiplexed proteomics. We defined the host response to infection at both the proteome and phospho-proteome levels. The proteome showed an increase in the immune response and a strong repression of several mitochondrial proteins. Complementing the proteome studies, the phospho-proteomic survey found an abundance of phospho-site alterations in plasma membrane and cytoskeletal proteins. Bioinformatic analysis of kinase activity provided substantial evidence for the activation of NDRG2 and JNK/p38 kinases during Chagas disease. A significant activation of DYRK2 and AMPKA2 and the inhibition of casein family kinases were also predicted. We concluded our analyses by linking the diseased heart proteome profile to known therapeutic interventions, uncovering a potential to target mitochondrial proteins, secreted immune effectors and core kinases for the treatment of chronic Chagas disease. Together, this study provides molecular insight into host proteome and phospho-proteome responses to T. cruzi infection in the heart for the first time, highlighting pathways that can be further validated for functional contributions to disease and suitability as drug targets.
Collapse
Affiliation(s)
- Jacob M. Wozniak
- Skaggs School of Pharmacy and Pharmaceutical Sciences; University of California San Diego; La Jolla, CA, United States of America
- Department of Pharmacology; University of California San Diego; La Jolla, CA, United States of America
| | - Tatiana Araújo Silva
- Cellular Ultrastructure Laboratory; Oswaldo Cruz Institute, FIOCRUZ; Rio de Janeiro, RJ, Brazil
| | - Diane Thomas
- Skaggs School of Pharmacy and Pharmaceutical Sciences; University of California San Diego; La Jolla, CA, United States of America
| | - Jair L. Siqueira-Neto
- Skaggs School of Pharmacy and Pharmaceutical Sciences; University of California San Diego; La Jolla, CA, United States of America
| | - James H. McKerrow
- Skaggs School of Pharmacy and Pharmaceutical Sciences; University of California San Diego; La Jolla, CA, United States of America
| | - David J. Gonzalez
- Skaggs School of Pharmacy and Pharmaceutical Sciences; University of California San Diego; La Jolla, CA, United States of America
- Department of Pharmacology; University of California San Diego; La Jolla, CA, United States of America
- * E-mail: (DJG); (CMC)
| | - Claudia M. Calvet
- Skaggs School of Pharmacy and Pharmaceutical Sciences; University of California San Diego; La Jolla, CA, United States of America
- Cellular Ultrastructure Laboratory; Oswaldo Cruz Institute, FIOCRUZ; Rio de Janeiro, RJ, Brazil
- * E-mail: (DJG); (CMC)
| |
Collapse
|
24
|
Cruz-Saavedra L, Muñoz M, Patiño LH, Vallejo GA, Guhl F, Ramírez JD. Slight temperature changes cause rapid transcriptomic responses in Trypanosoma cruzi metacyclic trypomastigotes. Parasit Vectors 2020; 13:255. [PMID: 32410662 PMCID: PMC7226949 DOI: 10.1186/s13071-020-04125-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 05/08/2020] [Indexed: 02/06/2023] Open
Abstract
Background Severe changes in temperature can affect the behavior and ecology of some infectious agents. Trypanosoma cruzi is a protozoan that causes Chagas disease. This parasite has high genetic variability and can be divided into six discrete typing units (DTUs). Trypanosoma cruzi also has a complex life-cycle, which includes the process of metacyclogenesis when non-infective epimastigote forms are differentiated into infective metacyclic trypomastigotes (MT). Studies in triatomines have shown that changes in temperature also affect the number and viability of MT. Methods The objective of this study was to evaluate how temperature affects the transcriptional profiles of T. cruzi I and II (TcI and TcII) MT by exposing parasites to two temperatures (27 °C and 28 °C) and comparing those to normal culture conditions at 26 °C. Subsequently, RNA-seq was conducted and differentially expressed genes were quantified and associated to metabolic pathways. Results A statistically significant difference was observed in the number of MT between the temperatures evaluated and the control, TcII DTU was not strongly affected to exposure to high temperatures compared to TcI. Similar results were found when we analyzed gene expression in this DTU, with the greatest number of differentially expressed genes being observed at 28 °C, which could indicate a dysregulation of different signaling pathways under this temperature. Chromosome analysis indicated that chromosome 1 harbored the highest number of changes for both DTUs for all thermal treatments. Finally, gene ontology (GO) analyses showed a decrease in the coding RNAs involved in the regulation of processes related to the metabolism of lipids and carbohydrates, the evasion of oxidative stress, and proteolysis and phosphorylation processes, and a decrease in RNAs coding to ribosomal proteins in TcI and TcII, along with an increase in the expression of surface metalloprotease GP63 in TcII. Conclusions Slight temperature shifts lead to increased cell death of metacyclic trypomastigotes because of the deregulation of gene expression of different processes essential for the TcI and TcII DTUs of T. cruzi.![]()
Collapse
Affiliation(s)
- Lissa Cruz-Saavedra
- Grupo de Investigaciones Microbiológicas-UR (GIMUR), Departamento de Biología, Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Marina Muñoz
- Grupo de Investigaciones Microbiológicas-UR (GIMUR), Departamento de Biología, Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Luz Helena Patiño
- Grupo de Investigaciones Microbiológicas-UR (GIMUR), Departamento de Biología, Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Gustavo A Vallejo
- Laboratorio de Investigaciones en Parasitología Tropical, Facultad de Ciencias, Universidad del Tolima, Ibagué, Colombia
| | - Felipe Guhl
- Centro de Investigaciones en Microbiología y Parasitología Tropical (CIMPAT), Facultad de Ciencias, Universidad de Los Andes, Bogotá, Colombia
| | - Juan David Ramírez
- Grupo de Investigaciones Microbiológicas-UR (GIMUR), Departamento de Biología, Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia.
| |
Collapse
|
25
|
Silva TA, Ferreira LFDC, Pereira MCDS, Calvet CM. Differential Role of TGF-β in Extracellular Matrix Regulation During Trypanosoma cruzi-Host Cell Interaction. Int J Mol Sci 2019; 20:E4836. [PMID: 31569452 PMCID: PMC6801917 DOI: 10.3390/ijms20194836] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 08/03/2019] [Accepted: 08/07/2019] [Indexed: 12/19/2022] Open
Abstract
Transforming growth factor beta (TGF-β) is a determinant for inflammation and fibrosis in cardiac and skeletal muscle in Chagas disease. To determine its regulatory mechanisms, we investigated the response of Trypanosoma cruzi-infected cardiomyocytes (CM), cardiac fibroblasts (CF), and L6E9 skeletal myoblasts to TGF-β. Cultures of CM, CF, and L6E9 were infected with T. cruzi (Y strain) and treated with TGF-β (1-10 ng/mL, 1 h or 48 h). Fibronectin (FN) distribution was analyzed by immunofluorescence and Western blot (WB). Phosphorylated SMAD2 (PS2), phospho-p38 (p-p38), and phospho-c-Jun (p-c-Jun) signaling were evaluated by WB. CF and L6E9 showed an increase in FN from 1 ng/mL of TGF-β, while CM displayed FN modulation only after 10 ng/mL treatment. CF and L6E9 showed higher PS2 levels than CM, while p38 was less stimulated in CF than CM and L6E9. T. cruzi infection resulted in localized FN disorganization in CF and L6E9. T. cruzi induced an increase in FN in CF cultures, mainly in uninfected cells. Infected CF cultures treated with TGF-β showed a reduction in PS2 and an increase in p-p38 and p-c-Jun levels. Our data suggest that p38 and c-Jun pathways may be participating in the fibrosis regulatory process mediated by TGF-β after T. cruzi infection.
Collapse
Affiliation(s)
- Tatiana Araújo Silva
- Cellular Ultrastructure Laboratory, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro 21040-360, Brazil.
| | | | | | - Claudia Magalhães Calvet
- Cellular Ultrastructure Laboratory, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro 21040-360, Brazil.
| |
Collapse
|
26
|
Liu Q, Zhu LJ, Waaga-Gasser AM, Ding Y, Cao M, Jadhav SJ, Kirollos S, Shekar PS, Padera RF, Chang YC, Xu X, Zeisberg EM, Charytan DM, Hsiao LL. The axis of local cardiac endogenous Klotho-TGF-β1-Wnt signaling mediates cardiac fibrosis in human. J Mol Cell Cardiol 2019; 136:113-124. [PMID: 31520610 DOI: 10.1016/j.yjmcc.2019.09.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 08/31/2019] [Accepted: 09/09/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND Cardiovascular fibrosis is a major contributor to cardiovascular disease, the primary cause of death in patients with chronic kidney disease (CKD). We previously reported expression of endogenous Klotho in human arteries, and that CKD is a state of Klotho deficiency, resulting in vascular calcification, but myocardial expression of Klotho is poorly understood. This study aimed to further clarify endogenous Klotho's functional roles in cardiac fibrosis in patients with underlying CKD. METHODS AND RESULTS Human atrial appendage specimens were collected during cardiac surgery from individuals with or without CKD. Cardiac fibrosis was quantified using trichrome staining. For endogenous Klotho functional studies, primary human cardiomyocytes (HCMs) were treated with uremic serum from CKD patients or recombinant human TGF-β1. The effects of endogenous Klotho in HCMs were studied using Klotho-siRNA and Klotho-plasmid transfection. Both gene and protein expression of endogenous Klotho are found in human heart, but decreased Klotho expression is clearly associated with the degree of cardiac fibrosis in CKD patients. Moreover, we show that endogenous Klotho is expressed by HCMs and cardiac fibroblasts (HCFs) but that HCM expression is suppressed by uremic serum or TGF-β1. Klotho knockdown or overexpression aggravates or mitigates TGF-β1-induced fibrosis and canonical Wnt signaling in HCMs, respectively. Furthermore, co-culture of HCMs with HCFs increases TGF-β1-induced fibrogenic proteins in HCFs, but overexpression of endogenous Klotho in HCMs mitigates this effect, suggesting functional crosstalk between HCMs and HCFs. CONCLUSIONS Our data from analysis of human hearts as well as functional in vitro studies strongly suggests that the loss of cardiac endogenous Klotho in CKD patients, specifically in cardiomyocytes, facilitates intensified TGF-β1 signaling which enables more vigorous cardiac fibrosis through upregulated Wnt signaling. Upregulation of endogenous Klotho inhibits pathogenic Wnt/β-catenin signaling and may offer a novel strategy for prevention and treatment of cardiac fibrosis in CKD patients.
Collapse
Affiliation(s)
- Qinghua Liu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Lang-Jing Zhu
- Department of Nephrology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China; Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ana Maria Waaga-Gasser
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yan Ding
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Minghua Cao
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Shreyas J Jadhav
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Sandra Kirollos
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Prem S Shekar
- Division of Cardiac Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Robert F Padera
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yu-Chun Chang
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Xingbo Xu
- Department of Cardiology and Pneumology, University Medical Center of Göttingen, Georg-August University, Göttingen, Lower Saxony 37075, Germany; German Centre for Cardiovascular Research (DZHK), Göttingen, Lower Saxony 37075, Germany
| | - Elisabeth M Zeisberg
- Department of Cardiology and Pneumology, University Medical Center of Göttingen, Georg-August University, Göttingen, Lower Saxony 37075, Germany; German Centre for Cardiovascular Research (DZHK), Göttingen, Lower Saxony 37075, Germany; Nephrology Division, Langone Medical Center, New York University, New York, NY 10016, USA
| | - David M Charytan
- Nephrology Division, Langone Medical Center, New York University, New York, NY 10016, USA.
| | - Li-Li Hsiao
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
27
|
Oliveira AER, Grazielle-Silva V, Ferreira LRP, Teixeira SMR. Close encounters between Trypanosoma cruzi and the host mammalian cell: Lessons from genome-wide expression studies. Genomics 2019; 112:990-997. [PMID: 31229555 DOI: 10.1016/j.ygeno.2019.06.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 04/22/2019] [Accepted: 06/15/2019] [Indexed: 12/15/2022]
Abstract
Trypanosoma cruzi is the etiologic agent of Chagas disease, a life-threatening disease that affects different tissues. Within its mammalian host, T. cruzi develops molecular strategies for successful invasion of different cell types and adaptation to the intracellular environment. Conversely, the host cell responds to the infection by activating intracellular pathways to control parasite replication. Here, we reviewed genome-wide expression studies based on microarray and RNA-seq data from both parasite and host genes generated from animal models of infection as well as from Chagas disease patients. As expected, analyses of T. cruzi genes highlighted changes related to parasite energy metabolism and cell surface molecules, whereas host cell transcriptome emphasized the role of immune response genes. Besides allowing a better understanding of mechanisms behind the pathogenesis of Chagas disease, these studies provide essential information for the development of new therapies as well as biomarkers for diagnosis and assessment of disease progression.
Collapse
Affiliation(s)
- Antonio Edson R Oliveira
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Viviane Grazielle-Silva
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ludmila R P Ferreira
- Departamento de Morfologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Santuza M R Teixeira
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
28
|
Shen J, Xing W, Gong F, Wang W, Yan Y, Zhang Y, Xie C, Fu S. MiR-150-5p retards the progression of myocardial fibrosis by targeting EGR1. Cell Cycle 2019; 18:1335-1348. [PMID: 31122130 DOI: 10.1080/15384101.2019.1617614] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
To investigate the differential expression of microRNA-150-5p (miR-150-5p) and early growth response 1 (EGR1) in myocardial fibrosis (MF) cells, and determine the effect between miR-150-5p and EGR1 on MF. Human MF cells were generated via Trypanosoma cruzi (T. cruzi) infection, a mouse model of MF was generated via angiotensin II. The expression levels of miR-150-5p and EGR1 were detected by quantitative real-time polymerase chain reaction (qRT-PCR) and western blot assay. The correlation between miR-150-5p and EGR1 was confirmed by a luciferase reporter assay. The viability, proliferation, and apoptotic rate were detected by cell counting kit-8 (CCK-8), colony-formation and flow cytometry assays. Hematoxylin-eosin (HE) staining and Masson staining visualized the degree of MF. Echocardiography was performed to obtain the levels of left ventricle fractional shortening (LVFS) and left ventricle ejection fraction (LVEF), computer algorithms and a videographics program were used to obtain the levels of left ventricular systolic pressure (LVSP), left ventricular end diastolic pressure (LVEDP) and ±left ventricular dp/dt maximum (LV dp/dtmax). We found that the expression of miR-150-5p in MF cells was lower than normal cardiomyocytes, while the expression level of EGR1 in MF cells were higher than normal cardiomyocytes. Cell experiments demonstrated that EGR1 and miR-150-5p could influence the development of MF, and the expression of EGR1 in cardiomyocytes was regulated by miR-150-5p directly. Lastly, we confirmed that sh-Egr1 would decrease the severity of MF, while miR-150-5p antagomir could aggravate MF. Our results illustrate the mechanism of MF development, and provide a potential target for MF treatment.
Collapse
Affiliation(s)
- Jie Shen
- a Department of Cardiology , Children's Hospital, Zhejiang University School of Medicine , Hangzhou , Zhejiang , PR China
| | - Wanhong Xing
- b Children's Heart Center , Sichuan Provincial Hospital for Women and Children , Chengdu , Sichuan , PR China
| | - Fangqi Gong
- a Department of Cardiology , Children's Hospital, Zhejiang University School of Medicine , Hangzhou , Zhejiang , PR China
| | - Wei Wang
- a Department of Cardiology , Children's Hospital, Zhejiang University School of Medicine , Hangzhou , Zhejiang , PR China
| | - Yufeng Yan
- c Trainning Center of Medical Experiments, School of Basic Medical Sciences , Fudan University , Shanghai , PR China
| | - Yiying Zhang
- a Department of Cardiology , Children's Hospital, Zhejiang University School of Medicine , Hangzhou , Zhejiang , PR China
| | - Chunhong Xie
- a Department of Cardiology , Children's Hospital, Zhejiang University School of Medicine , Hangzhou , Zhejiang , PR China
| | - Songling Fu
- a Department of Cardiology , Children's Hospital, Zhejiang University School of Medicine , Hangzhou , Zhejiang , PR China
| |
Collapse
|
29
|
Suman S, Rachakonda G, Mandape SN, Sakhare SS, Villalta F, Pratap S, Lima MF, Nde PN. Phospho-proteomic analysis of primary human colon epithelial cells during the early Trypanosoma cruzi infection phase. PLoS Negl Trop Dis 2018; 12:e0006792. [PMID: 30222739 PMCID: PMC6160231 DOI: 10.1371/journal.pntd.0006792] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 09/27/2018] [Accepted: 08/27/2018] [Indexed: 12/11/2022] Open
Abstract
The protozoan parasite Trypanosoma cruzi, the causative agent of Chagas disease, causes severe morbidity and mortality in afflicted individuals. About 30% of T. cruzi-infected individuals present with cardiac, gastrointestinal tract, and/or neurological disorders. Megacolon, one of the major pathologies of Chagas disease, is accompanied by gastrointestinal motility disorders. The molecular mechanism of T. cruzi-mediated megacolon in Chagas disease is currently unknown. To decipher the molecular mechanism of T. cruzi-induced alteration in the colon during the early infection phase, we exposed primary human colonic epithelial cells (HCoEpiC) to invasive T. cruzi trypomastigotes at multiple time points to determine changes in the phosphoprotein networks in the cells following infection using proteome profiler Human phospho-kinase arrays. We found significant changes in the phosphorylation pattern that can mediate cellular deregulations in colonic epithelial cells after infection. We detected a significant increase in the levels of phosphorylated heat shock protein (p-HSP) 27 and transcription factors that regulate various cellular functions, including c-Jun and CREB. Our study confirmed significant upregulation of phospho (p-) Akt S473, p-JNK, which may directly or indirectly modulate CREB and c-Jun phosphorylation, respectively. We also observed increased levels of phosphorylated CREB and c-Jun in the nucleus. Furthermore, we found that p-c-Jun and p-CREB co-localized in the nucleus at 180 minutes post infection, with a maximum Pearson correlation coefficient of 0.76±0.02. Increased p-c-Jun and p-CREB have been linked to inflammatory and profibrotic responses. T. cruzi infection of HCoEpiC induces an increased expression of thrombospondin-1 (TSP-1), which is fibrogenic at elevated levels. We also found that T. cruzi infection modulates the expression of NF-kB and JAK2-STAT1 signaling molecules which can increase pro-inflammatory flux. Bioinformatics analysis of the phosphoprotein networks derived using the phospho-protein data serves as a blueprint for T. cruzi-mediated cellular transformation of primary human colonic cells during the early phase of T. cruzi infection.
Collapse
Affiliation(s)
- Shankar Suman
- Department of Microbiology and Immunology, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Girish Rachakonda
- Department of Microbiology and Immunology, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Sammed N. Mandape
- Department of Microbiology and Immunology, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Shruti S. Sakhare
- Department of Microbiology and Immunology, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Fernando Villalta
- Department of Microbiology and Immunology, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Siddharth Pratap
- School of Graduate Studies and Research, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Maria F. Lima
- School of Graduate Studies and Research, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Pius N. Nde
- Department of Microbiology and Immunology, Meharry Medical College, Nashville, Tennessee, United States of America
| |
Collapse
|
30
|
Libisch MG, Faral-Tello P, Garg NJ, Radi R, Piacenza L, Robello C. Early Trypanosoma cruzi Infection Triggers mTORC1-Mediated Respiration Increase and Mitochondrial Biogenesis in Human Primary Cardiomyocytes. Front Microbiol 2018; 9:1889. [PMID: 30166980 PMCID: PMC6106620 DOI: 10.3389/fmicb.2018.01889] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 07/27/2018] [Indexed: 12/31/2022] Open
Abstract
Chagasic chronic cardiomyopathy is one of the most frequent and severe manifestations of Chagas disease, caused by the parasite Trypanosoma cruzi. The pathogenic and biochemical mechanisms responsible for cardiac lesions remain not completely understood, although it is clear that hypertrophy and subsequent heart dilatation is in part caused by the direct infection of cardiomyocytes. In this work, we evaluated the initial response of human cardiomyocytes to T. cruzi infection by transcriptomic profiling. Immediately after infection, cardiomyocytes dramatically change their gene expression patterns, up regulating most of the genes encoding for respiratory chain, oxidative phosphorylation and protein synthesis. We found that these changes correlate with an increase in basal and maximal respiration, as well as in spare respiratory capacity, which is accompanied by mitochondrial biogenesis pgc-1α independent. We also demonstrate that these changes are mediated by mTORC1 and reversed by rapamycin, resembling the molecular mechanisms described for the non-chagasic hypertrophic cardiomyopathy. The results of the present work identify that early during infection, the activation of mTORC1, mitochondrial biogenesis and improvement in oxidative phosphorylation are key biochemical changes that provide new insights into the host response to parasite infection and the pathogenesis of chronic chagasic cardiomyopathy. The finding that this phenotype can be reversed opens a new perspective in the treatment of Chagas disease, through the identification of host targets, and the use of combined parasite and host targeted therapies, in order to prevent chagasic cardiomyopathy.
Collapse
Affiliation(s)
- M Gabriela Libisch
- Laboratory of Host-Pathogen Interactions-UBM, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Paula Faral-Tello
- Laboratory of Host-Pathogen Interactions-UBM, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Nisha J Garg
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, TX, United States
| | - Rafael Radi
- Departamento de Bioquímica, Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Lucía Piacenza
- Departamento de Bioquímica, Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Carlos Robello
- Laboratory of Host-Pathogen Interactions-UBM, Institut Pasteur de Montevideo, Montevideo, Uruguay.,Departamento de Bioquímica, Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
31
|
Pathogens and Their Effect on Exosome Biogenesis and Composition. Biomedicines 2018; 6:biomedicines6030079. [PMID: 30041409 PMCID: PMC6164629 DOI: 10.3390/biomedicines6030079] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 07/17/2018] [Accepted: 07/17/2018] [Indexed: 12/12/2022] Open
Abstract
Exosomes are nanosized membrane microvesicles (30⁻100 nm) that have the capability to communicate intercellularly and transport cell components (i.e., miRNA, mRNA, proteins and DNA). Exosomes are found in nearly every cell type (i.e., mast cells, dendritic, tumor, and macrophages). There have been many studies that have shown the importance of exosome function as well as their unique packaging and targeting abilities. These characteristics make exosomes ideal candidates to act as biomarkers and therapeutics for disease. We will discuss the biogenesis, composition, and relationship of exosomes with non-viral microbial infections including gram-negative bacteria, gram-positive bacteria, Leishmania and Trypanosoma cruzi.
Collapse
|
32
|
Ferreira BL, Ferreira ÉR, de Brito MV, Salu BR, Oliva MLV, Mortara RA, Orikaza CM. BALB/c and C57BL/6 Mice Cytokine Responses to Trypanosoma cruzi Infection Are Independent of Parasite Strain Infectivity. Front Microbiol 2018; 9:553. [PMID: 29662478 PMCID: PMC5890190 DOI: 10.3389/fmicb.2018.00553] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 03/12/2018] [Indexed: 11/13/2022] Open
Abstract
Trypanosoma cruzi is the etiologic agent of Chagas’ disease, which affects 6–7 million people worldwide. Different strains of T. cruzi present specific genotypic and phenotypic characteristics that affect the host–pathogen interactions, and thus, the parasite has been classified into six groups (TcI to TcVI). T. cruzi infection presents two clinical phases, acute and chronic, both with distinct characteristics and important participation by the immune system. However, the specific contributions of parasite and host factors in the disease phases are not yet fully understood. The murine model for Chagas’ disease is well-established and reproduces important features of the human infection, providing an experimental basis for the study of host lineages and parasite strains. Thus, we evaluated acute and chronic infection by the G (TcI) and CL (TcVI) strains of T. cruzi, which have distinct tropisms and infectivity, in two inbred mice lineages (C57BL/6 and BALB/c) that display variable degrees of susceptibility to different T. cruzi strains. Analysis of the parasite loads in host tissues by qPCR showed that CL strain established an infection faster than the G strain; at the same time, the response in BALB/c mice, although diverse in terms of cytokine secretion, was initiated earlier than that in C57BL/6 mice. At the parasitemia peak in the acute phase, we observed, either by confocal microscopy or by qPCR, that the infection was disseminated in all groups analyzed, with some differences concerning parasite tropism; at this point, all animals responded to infection by increasing the serum concentrations of cytokines. However, BALB/c mice seemed to better regulate the immune response than C57BL/6 mice. Indeed, in the chronic phase, C57BL/6 mice still presented exacerbated cytokine and chemokine responses. In summary, our results indicate that in these experimental models, the deregulation of immune response that is typical of chronic Chagas’ disease may be due to control loss over pro- and anti-inflammatory cytokines early in the acute phase of the disease, depending primarily on the host background rather than the parasite strain.
Collapse
Affiliation(s)
- Bianca L Ferreira
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Éden R Ferreira
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Marlon V de Brito
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Bruno R Salu
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Maria L V Oliva
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Renato A Mortara
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Cristina M Orikaza
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
33
|
Coelho LL, Pereira IR, Pereira MCDS, Mesquita L, Lannes-Vieira J, Adesse D, Garzoni LR. Trypanosoma cruzi activates mouse cardiac fibroblasts in vitro leading to fibroblast-myofibroblast transition and increase in expression of extracellular matrix proteins. Parasit Vectors 2018; 11:72. [PMID: 29382361 PMCID: PMC5791182 DOI: 10.1186/s13071-018-2614-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 01/02/2018] [Indexed: 01/05/2023] Open
Abstract
Background Cardiac fibrosis is a consequence of chronic chagasic cardiomyopathy (CCC). In other cardiovascular diseases, the protagonist role of fibroblasts in cardiac fibrosis is well established. However, the role of cardiac fibroblasts (CFs) in fibrosis during the CCC is not clear. Here, our aim was to investigate the effect of Trypanosoma cruzi, the etiological agent of Chagas disease on CFs activation. Methods Cardiac fibroblasts were purified from primary cultures of mouse embryo cardiac cells. After two passages, cells were infected with T. cruzi (Y strain) and analyzed at different times for determination of infectivity, activation and production of extracellular matrix components (fibronectin, laminin and collagen IV) by immunofluorescence and western blot. Results At second passage, cultures were enriched in CFs (95% of fibroblasts and 5% of cardiomyocytes), as revealed by presence of alpha-smooth muscle actin (α-SMA) and discoidin domain receptor 2 (DDR2) and absence of sarcomeric tropomyosin (ST) protein expression. Trypanosoma cruzi infection induced fibroblast-myofibroblast transition, with increased expression of α-SMA after 6 and 24 h post-infection (hpi). Fibronectin was increased at 6, 24 and 48 hpi, laminin was increased at 6 and 24 hpi and collagen IV was increased at 6 hpi. Conclusions Our results showed that T. cruzi activates CFs, inducing activation and exacerbates ECM production. Furthermore, our data raise the possibility of the involvement of CFs in heart fibrosis during Chagas disease.
Collapse
Affiliation(s)
- Laura Lacerda Coelho
- Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Av. Brasil 4365, Pavilhão Cardoso Fontes, 2° andar, Rio de Janeiro RJ, 20045-900, Brazil
| | - Isabela Resende Pereira
- Laboratório de Biologia das Interações, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Av. Brasil 4365, Pavilhão Cardoso Fontes, 2° andar, Rio de Janeiro RJ, 20045-900, Brazil
| | - Mirian Claudia de Souza Pereira
- Laboratório de Ultraestrutura Celular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Av. Brasil 4365, Pavilhão Carlos Chagas sala 308, Rio de Janeiro RJ, 20045-900, Brazil
| | - Liliane Mesquita
- Laboratório de Ultraestrutura Celular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Av. Brasil 4365, Pavilhão Carlos Chagas sala 308, Rio de Janeiro RJ, 20045-900, Brazil
| | - Joseli Lannes-Vieira
- Laboratório de Biologia das Interações, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Av. Brasil 4365, Pavilhão Cardoso Fontes, 2° andar, Rio de Janeiro RJ, 20045-900, Brazil
| | - Daniel Adesse
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Av. Brasil 4365, Pavilhão Carlos Chagas, sala 307, Rio de Janeiro RJ, 20045-900, Brazil
| | - Luciana Ribeiro Garzoni
- Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Av. Brasil 4365, Pavilhão Cardoso Fontes, 2° andar, Rio de Janeiro RJ, 20045-900, Brazil.
| |
Collapse
|
34
|
Wen JJ, Porter C, Garg NJ. Inhibition of NFE2L2-Antioxidant Response Element Pathway by Mitochondrial Reactive Oxygen Species Contributes to Development of Cardiomyopathy and Left Ventricular Dysfunction in Chagas Disease. Antioxid Redox Signal 2017; 27:550-566. [PMID: 28132522 PMCID: PMC5567598 DOI: 10.1089/ars.2016.6831] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
AIMS We investigated the effects of mitochondrial reactive oxygen species (mtROS) on nuclear factor (erythroid 2)-like 2 (NFE2L2) transcription factor activity during Trypanosoma cruzi (Tc) infection and determined whether enhancing the mtROS scavenging capacity preserved the heart function in Chagas disease. RESULTS C57BL/6 wild type (WT, female) mice infected with Tc exhibited myocardial loss of mitochondrial membrane potential, complex II (CII)-driven coupled respiration, and ninefold increase in mtROS production. In vitro and in vivo studies showed that Tc infection resulted in an ROS-dependent decline in the expression, nuclear translocation, antioxidant response element (ARE) binding, and activity of NFE2L2, and 35-99% decline in antioxidants' (gamma-glutamyl cysteine synthase [γGCS], heme oxygenase-1 [HO1], glutamate-cysteine ligase modifier subunit [GCLM], thioredoxin (Trx), glutathione S transferase [GST], and NAD(P)H dehydrogenase, quinone 1 [NQO1]) expression. An increase in myocardial and mitochondrial oxidative adducts, myocardial interventricular septum thickness, and left ventricle (LV) mass, a decline in LV posterior wall thickness, and disproportionate synthesis of collagens (COLI/COLIII), αSMA, and SM22α were noted in WT.Tc mice. Overexpression of manganese superoxide dismutase (MnSOD) in cultured cells (HeLa or cardiomyocytes) and MnSODtg mice preserved the NFE2L2 transcriptional activity and antioxidant/oxidant balance, and cardiac oxidative and fibrotic pathology were significantly decreased in MnSODtg.Tc mice. Importantly, echocardiography finding of a decline in LV systolic (stroke volume, cardiac output, ejection fraction) and diastolic (early/late peak filling ratio, myocardial performance index) function in WT.Tc mice was abolished in MnSODtg.Tc mice. Innovation and Conclusion: The mtROS inhibition of NFE2L2/ARE pathway constitutes a key mechanism in signaling the fibrotic gene expression and evolution of chronic cardiomyopathy. Preserving the NFE2L2 activity arrested the mitochondrial and cardiac oxidative stress, cardiac fibrosis, and heart failure in Chagas disease. Antioxid. Redox Signal. 27, 550-566.
Collapse
Affiliation(s)
- Jake Jianjun Wen
- 1 Department of Microbiology and Immunology, University of Texas Medical Branch (UTMB) , Galveston, Texas
| | - Craig Porter
- 2 Metabolism Unit, Shriners Hospital for Children , Galveston, Texas.,3 Department of Surgery, University of Texas Medical Branch (UTMB) , Galveston, Texas
| | - Nisha Jain Garg
- 1 Department of Microbiology and Immunology, University of Texas Medical Branch (UTMB) , Galveston, Texas.,4 Department of Pathology, University of Texas Medical Branch (UTMB) , Galveston, Texas.,5 Institute for Human Infections and Immunity, University of Texas Medical Branch (UTMB) , Galveston, Texas
| |
Collapse
|
35
|
Genetic Adjuvantation of a Cell-Based Therapeutic Vaccine for Amelioration of Chagasic Cardiomyopathy. Infect Immun 2017; 85:IAI.00127-17. [PMID: 28674032 DOI: 10.1128/iai.00127-17] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 06/21/2017] [Indexed: 12/19/2022] Open
Abstract
Chagas disease, caused by infection with the protozoan parasite Trypanosoma cruzi, is a leading cause of heart disease ("chagasic cardiomyopathy") in Latin America, disproportionately affecting people in resource-poor areas. The efficacy of currently approved pharmaceutical treatments is limited mainly to acute infection, and there are no effective treatments for the chronic phase of the disease. Preclinical models of Chagas disease have demonstrated that antigen-specific CD8+ gamma interferon (IFN-γ)-positive T-cell responses are essential for reducing parasite burdens, increasing survival, and decreasing cardiac pathology in both the acute and chronic phases of Chagas disease. In the present study, we developed a genetically adjuvanted, dendritic cell-based immunotherapeutic for acute Chagas disease in an attempt to delay or prevent the cardiac complications that eventually result from chronic T. cruzi infection. Dendritic cells transduced with the adjuvant, an adenoviral vector encoding a dominant negative isoform of Src homology region 2 domain-containing tyrosine phosphatase 1 (SHP-1) along with the T. cruzi Tc24 antigen and trans-sialidase antigen 1 (TSA1), induced significant numbers of antigen-specific CD8+ IFN-γ-positive cells following injection into BALB/c mice. A vaccine platform transduced with the adenoviral vector and loaded in tandem with the recombinant protein reduced parasite burdens by 76% to >99% in comparison to a variety of different controls and significantly reduced cardiac pathology in a BALB/c mouse model of live Chagas disease. Although no statistical differences in overall survival rates among cohorts were observed, the data suggest that immunotherapeutic strategies for the treatment of acute Chagas disease are feasible and that this approach may warrant further study.
Collapse
|
36
|
Wen JJ, Wan X, Thacker J, Garg NJ. Chemotherapeutic efficacy of phosphodiesterase inhibitors in chagasic cardiomyopathy. JACC Basic Transl Sci 2016; 1:235-250. [PMID: 27747306 PMCID: PMC5065248 DOI: 10.1016/j.jacbts.2016.04.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Molecular mechanisms of Trypanosoma cruzi (Tc)-induced Chagasic cardiomyopathy (CCM) are not well understood. The NO-cGMP-PKG1α pathway maintains cardiac homeostasis and inotropy and may be disturbed due to phosphodiesterase (PDE5)-mediated cGMP catabolism in CCM. To test this, C57BL/6 mice were infected with T. cruzi, and after the control of acute parasitemia (∼45 days post-infection), given sildenafil (SIL) (1 mg/kg) treatment for 3 weeks that ended long before the chronic disease phase (∼150 days post-infection). The PDE5 was increased and cGMP/PKG activity was decreased in chagasic myocardium. Transthoracic echocardiography revealed left ventricular (LV) systolic function, that is, stroke volume, cardiac output, and ejection fraction, was significantly decreased in chagasic mice. SIL treatment resulted in normal levels of PDE5 and cGMP/PKG activity and preserved the LV function. The cardioprotective effects of SIL were provided through inhibition of cardiac collagenosis and chronic inflammation that otherwise were pronounced in CCM. Further, SIL treatment restored the mitochondrial DNA–encoded gene expression, complex I–dependent (but not complex II–dependent) ADP-coupled respiration, and oxidant/antioxidant balance in chagasic myocardium. In vitro studies in cardiomyocytes verified that SIL conserved the redox metabolic state and cellular health via maintaining the antioxidant status that otherwise was compromised in response to T. cruzi infection. We conclude that SIL therapy was useful in controlling the LV dysfunction and chronic pathology in CCM. Mice infected with T. cruzi control acute parasitemia but develop chronic chagasic cardiomyopathy. Treatment with SIL (a phosphodiesterase inhibitor) during a therapeutic window of indeterminate phase provided powerful cardioprotective effects against chronic development of cardiomyopathy and LV dysfunction. SIL normalized the cGMP-dependent protein kinase activity and mitochondrial oxidative metabolism, and established the oxidant/antioxidant balance in chagasic myocardium. SIL prevented the oxidative/inflammatory adducts that precipitate cardiomyocytes death and cardiac remodeling in CCM.
Collapse
Affiliation(s)
- Jian-Jun Wen
- Department of Microbiology and Immunology, University of Texas Medical Branch (UTMB), Galveston, Texas
| | - Xianxiu Wan
- Department of Microbiology and Immunology, University of Texas Medical Branch (UTMB), Galveston, Texas
| | - John Thacker
- Department of Microbiology and Immunology, University of Texas Medical Branch (UTMB), Galveston, Texas
| | - Nisha Jain Garg
- Department of Microbiology and Immunology, University of Texas Medical Branch (UTMB), Galveston, Texas; Department of Pathology, UTMB, Galveston, TX; Institute for Human Infections and Immunity, UTMB, Galveston, TX
| |
Collapse
|