1
|
Garcez EM, Gomes N, Moraes AS, Pogue R, Uenishi RH, Hecht M, Carvalho JL. Extracellular vesicles in the context of Chagas Disease - A systematic review. Acta Trop 2023; 242:106899. [PMID: 36935050 DOI: 10.1016/j.actatropica.2023.106899] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/13/2023] [Accepted: 03/16/2023] [Indexed: 03/19/2023]
Abstract
Extracellular vesicle (EVs) traffic is considered an important cellular communication process between cells that can be part of a single organism or belong to different living beings. The relevance of EV-mediated cellular communication is increasingly studied and appreciated, especially in relation to pathological conditions, including parasitic disorders, in which the EV release and uptake processes have been documented. In the context of Chagas Disease (CD), EVs have been explored, however, current data have not been systematically revised in order to provide an overview of the published literature and the main results obtained thus far. In this systematic review, 25 studies involving the investigation of EVs in CD were identified. The studies involved Trypanosoma cruzi (Tc)-derived EVs (Tc-EVs), as well as EVs derived from T. cruzi-infected mammalian cells-derived EVs, mainly isolated by ultracentrifugation and poorly characterized. The objectives of the identified studies included the characterization of the protein and RNA cargo of Tc-EVs, as well as investigation of EVs in parasitic infections and immune-related processes. Overall, our systematic review reveals that EVs play critical roles in several mechanisms related to the interaction between T. cruzi and mammalian hosts, their contribution to immune system evasion by the parasite, and to chronic inflammation in the host. Future studies will benefit from the consolidation of isolation and characterization methods, as well as the elucidation of the role of EVs in CD.
Collapse
Affiliation(s)
- Emãnuella Melgaço Garcez
- Multidisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasília, 70910-900, Brasília, DF, Brazil
| | - Nélio Gomes
- Multidisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasília, 70910-900, Brasília, DF, Brazil
| | - Aline Silva Moraes
- Multidisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasília, 70910-900, Brasília, DF, Brazil
| | - Robert Pogue
- Genomic Sciences and Biotechnology Program. Catholic University of Brasília, 71966-700, Brasília, DF, Brazil
| | - Rosa Harumi Uenishi
- Multidisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasília, 70910-900, Brasília, DF, Brazil
| | - Mariana Hecht
- Multidisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasília, 70910-900, Brasília, DF, Brazil
| | - Juliana Lott Carvalho
- Multidisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasília, 70910-900, Brasília, DF, Brazil; Genomic Sciences and Biotechnology Program. Catholic University of Brasília, 71966-700, Brasília, DF, Brazil.
| |
Collapse
|
2
|
Ferri G, Musikant D, Edreira MM. Host Cell Rap1b mediates cAMP-dependent invasion by Trypanosoma cruzi. PLoS Negl Trop Dis 2023; 17:e0011191. [PMID: 36897926 PMCID: PMC10032529 DOI: 10.1371/journal.pntd.0011191] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 03/22/2023] [Accepted: 02/22/2023] [Indexed: 03/11/2023] Open
Abstract
Trypanosoma cruzi cAMP-mediated invasion has long been described, however, the detailed mechanism of action of the pathway activated by this cyclic nucleotide still remains unknown. We have recently demonstrated a crucial role for Epac in the cAMP-mediated invasion of the host cell. In this work, we gathered evidence indicating that the cAMP/Epac pathway is activated in different cells lines. In accordance, data collected from pull-down experiments designed to identify only the active form of Rap1b (Rap1b-GTP), and infection assays using cells transfected with a constitutively active mutant of Rap1b (Rap1b-G12V), strongly suggest the participation of Rap1b as mediator of the pathway. In addition to the activation of this small GTPase, fluorescence microscopy allowed us to demonstrate the relocalization of Rap1b to the entry site of the parasite. Moreover, phospho-mimetic and non-phosphorylable mutants of Rap1b were used to demonstrate a PKA-dependent antagonistic effect on the pathway, by phosphorylation of Rap1b, and potentially of Epac. Finally, Western Blot analysis was used to determine the involvement of the MEK/ERK signalling downstream of cAMP/Epac/Rap1b-mediated invasion.
Collapse
Affiliation(s)
- Gabriel Ferri
- CONICET-Universidad de Buenos Aires, IQUIBICEN, Ciudad de Buenos Aires, Argentina
- Laboratorio de Biología Molecular de Trypanosomas, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos, Ciudad de Buenos Aires, Argentina
| | - Daniel Musikant
- CONICET-Universidad de Buenos Aires, IQUIBICEN, Ciudad de Buenos Aires, Argentina
- Laboratorio de Biología Molecular de Trypanosomas, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos, Ciudad de Buenos Aires, Argentina
| | - Martin M Edreira
- CONICET-Universidad de Buenos Aires, IQUIBICEN, Ciudad de Buenos Aires, Argentina
- Laboratorio de Biología Molecular de Trypanosomas, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos, Ciudad de Buenos Aires, Argentina
| |
Collapse
|
3
|
Depletion of Na+/H+ Exchanger Isoform 1 Increases the Host Cell Resistance to Trypanosoma cruzi Invasion. Pathogens 2022; 11:pathogens11111294. [DOI: 10.3390/pathogens11111294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 11/06/2022] Open
Abstract
Na+/H+ exchanger isoform 1 (NHE1), a member of a large family of integral membrane proteins, plays a role in regulating the cortical actin cytoskeleton. Trypanosoma cruzi, the agent of Chagas disease, depends on F-actin rearrangement and lysosome mobilization to invade host cells. To determine the involvement of NHE1 in T. cruzi metacyclic trypomastigote (MT) internalization, the effect of treatment in cells with NHE1 inhibitor amiloride or of NHE1 depletion was examined in human epithelial cells. MT invasion decreased in amiloride-treated and NHE1-depleted cells. The phosphorylation profile of diverse protein kinases, whose activation is associated with remodeling of actin fibers, was analyzed in amiloride-treated and NHE1-depleted cells. In amiloride-treated cells, the phosphorylation levels of protein kinase C (PKC), focal adhesion kinase (FAK) and Akt were similar to those of untreated cells, whereas those of extracellular signal-regulated protein kinases (ERK1/2) increased. In NHE1-deficient cells, with marked alteration in the actin cytoskeleton architecture and in lysosome distribution, the levels of phospho-PKC and phospho-FAK decreased, whereas those of phospho-Akt and phospho-ERK1/2 increased. These data indicate that NHE1 plays a role in MT invasion, by maintaining the activation status of diverse protein kinases in check and preventing the inappropriate F-actin arrangement that affects lysosome distribution.
Collapse
|
4
|
Guo Y, Dong HY, Zhou HC, Zhang ZS, Zhao Y, Zhang YJ. Mechanism of the Passage of Angiostrongylus cantonensis across the Final Host Blood-Brain Barrier Using the Next-Generation Sequencing. IRANIAN JOURNAL OF PARASITOLOGY 2021; 16:454-463. [PMID: 34630591 PMCID: PMC8476730 DOI: 10.18502/ijpa.v16i3.7099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 04/09/2021] [Indexed: 11/24/2022]
Abstract
Background: Multicellular parasites Angiostrogylus cantonensis larvae develop in the final host rat brain at the fourth stage (L4) and migrate to the lungs by the adult stage. The potential mechanism of its blood-brain barrier (BBB) passage remains unclear. Methods: By using Illumina Hiseq/Miseq sequencing, we obtained the transcriptomes of 3 groups of adult males and 3 groups of female of A. cantonensis to generate similarly expressed genes (SEGs) between 2 genders at the adult stage. Next 2 groups of L4 expressed genes were used to compared with SEGs to create differentially expressed genes (DEGs) between 2 life stages to unlock potential mechanism of BBB passage. Results: In total, we obtained 381 581 802 clean reads and 56 990 699 010 clean bases. Of these, 331 803 unigenes and 482 056 transcripts were successfully annotated. A total of 3 166 DEGs between L4 and adults SEGs were detected. Annotation of these DEGs showed 167 were down-regulated and 181 were up-regulated. Pathway analysis exhibited that calcium signaling pathway, the ECM−receptor interaction, focal adhesion, and cysteine and methionine metabolism were highly associated with DEGs. The function of these pathways might be related to BBB traversal, as well as neuro-regulation, interactions between parasite and host, environmental adaption. Conclusion: This study expanded the regulatory characteristics of the two important life stages of A. cantonensis. This information may provide a better appreciation of the biological features of the stages of the parasitic A. cantonensis.
Collapse
Affiliation(s)
- Yue Guo
- School of Medicine, Huzhou University, Huzhou Cent Hosp, Zhejiang, China.,Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, Huzhou University, Huzhou, Zhejiang, China
| | - Hai Yan Dong
- School of Medicine, Huzhou University, Huzhou Cent Hosp, Zhejiang, China.,Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, Huzhou University, Huzhou, Zhejiang, China
| | - Hong Chang Zhou
- School of Medicine, Huzhou University, Huzhou Cent Hosp, Zhejiang, China.,Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, Huzhou University, Huzhou, Zhejiang, China
| | - Zhong Shan Zhang
- School of Medicine, Huzhou University, Huzhou Cent Hosp, Zhejiang, China.,School of Life Sciences, Huzhou University, Zhejiang, China
| | - Yu Zhao
- School of Medicine, Huzhou University, Huzhou Cent Hosp, Zhejiang, China
| | - Yu Jie Zhang
- School of Medicine, Huzhou University, Huzhou Cent Hosp, Zhejiang, China
| |
Collapse
|
5
|
Onofre TS, Rodrigues JPF, Shio MT, Macedo S, Juliano MA, Yoshida N. Interaction of Trypanosoma cruzi Gp82 With Host Cell LAMP2 Induces Protein Kinase C Activation and Promotes Invasion. Front Cell Infect Microbiol 2021; 11:627888. [PMID: 33777840 PMCID: PMC7996063 DOI: 10.3389/fcimb.2021.627888] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 02/23/2021] [Indexed: 11/13/2022] Open
Abstract
The surface molecule gp82 of metacyclic trypomastigote (MT) forms of Trypanosoma cruzi, the protozoan parasite that causes Chagas disease, mediates the host cell invasion, a process critical for the establishment of infection. Gp82 is known to bind to the target cell in a receptor-dependent manner, triggering Ca2+ signal, actin cytoskeleton rearrangement and lysosome spreading. The host cell receptor for gp82 was recently identified as LAMP2, the major lysosome membrane-associated protein. To further clarify the mechanisms of MT invasion, we aimed in this study at identifying the LAMP2 domain that interacts with gp82 and investigated whether target cell PKC and ERK1/2, previously suggested to be implicated in MT invasion, are activated by gp82. Interaction of MT, or the recombinant gp82 (r-gp82), with human epithelial HeLa cells induced the activation of Ca2+-dependent PKC and ERK1/2. The LAMP2 sequence predicted to bind gp82 was mapped and the synthetic peptide based on that sequence inhibited MT invasion, impaired the binding of r-gp82 to HeLa cells, and blocked the PKC and ERK1/2 activation induced by r-gp82. Treatment of HeLa cells with specific inhibitor of focal adhesion kinase resulted in inhibition of r-gp82-induced PKC and ERK1/2 activation, as well as in alteration of the actin cytoskeleton architecture. PKC activation by r-gp82 was also impaired by treatment of HeLa cells with inhibitor of phospholipase C, which mediates the production of diacylglycerol, which activates PKC, and inositol 1,4,5-triphosphate that releases Ca2+ from intracellular stores. Taken together, our results indicate that recognition of MT gp82 by LAMP2 induces in the host cell the activation of phosholipase C, with generation of products that contribute for PKC activation and the downstream ERK1/2. This chain of events leads to the actin cytoskeleton disruption and lysosome spreading, promoting MT internalization.
Collapse
Affiliation(s)
- Thiago Souza Onofre
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - João Paulo Ferreira Rodrigues
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Marina Tiemi Shio
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Silene Macedo
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Maria Aparecida Juliano
- Departamento de Biofísica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Nobuko Yoshida
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
6
|
Nájera CA, Batista MF, Meneghelli I, Bahia D. Mixed signals - how Trypanosoma cruzi exploits host-cell communication and signaling to establish infection. J Cell Sci 2021; 134:134/5/jcs255687. [PMID: 33692153 DOI: 10.1242/jcs.255687] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Chagas disease (American trypanosomiasis) is a 'neglected' pathology that affects millions of people worldwide, mainly in Latin America. Trypanosoma cruzi, the causative agent, is an obligate intracellular parasite with a complex and diverse biology that infects several mammalian species, including humans. Because of genetic variability among strains and the presence of four biochemically and morphologically distinct parasite forms, the outcome of T. cruzi infection varies considerably depending on host cell type and parasite strain. During the initial contact, cellular communication is established by host-recognition-mediated responses, followed by parasite adherence and penetration. For this purpose, T. cruzi expresses a variety of proteins that modify the host cell, enabling it to safely reach the cytoplasm. After entry into the host cell, T. cruzi forms a transitory structure termed 'parasitophorous vacuole' (PV), followed by its cytoplasmic replication and differentiation after PV rupture, and subsequent invasion of other cells. The success of infection, maintenance and survival inside host cells is facilitated by the ability of T. cruzi to subvert various host signaling mechanisms. We focus in this Review on the various mechanisms that induce host cytoskeletal rearrangements, activation of autophagy-related proteins and crosstalk among major immune response regulators, as well as recent studies on the JAK-STAT pathway.
Collapse
Affiliation(s)
- Carlos Acides Nájera
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 486, Brazil
| | - Marina Ferreira Batista
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 486, Brazil
| | - Isabela Meneghelli
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 486, Brazil
| | - Diana Bahia
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 486, Brazil
| |
Collapse
|
7
|
Ferri G, Edreira MM. All Roads Lead to Cytosol: Trypanosoma cruzi Multi-Strategic Approach to Invasion. Front Cell Infect Microbiol 2021; 11:634793. [PMID: 33747982 PMCID: PMC7973469 DOI: 10.3389/fcimb.2021.634793] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 01/27/2021] [Indexed: 12/17/2022] Open
Abstract
T. cruzi has a complex life cycle involving four developmental stages namely, epimastigotes, metacyclic trypomastigotes, amastigotes and bloodstream trypomastigotes. Although trypomastigotes are the infective forms, extracellular amastigotes have also shown the ability to invade host cells. Both stages can invade a broad spectrum of host tissues, in fact, almost any nucleated cell can be the target of infection. To add complexity, the parasite presents high genetic variability with differential characteristics such as infectivity. In this review, we address the several strategies T. cruzi has developed to subvert the host cell signaling machinery in order to gain access to the host cell cytoplasm. Special attention is made to the numerous parasite/host protein interactions and to the set of signaling cascades activated during the formation of a parasite-containing vesicle, the parasitophorous vacuole, from which the parasite escapes to the cytosol, where differentiation and replication take place.
Collapse
Affiliation(s)
- Gabriel Ferri
- CONICET-Universidad de Buenos Aires, IQUIBICEN, Ciudad de Buenos Aires, Argentina
| | - Martin M Edreira
- CONICET-Universidad de Buenos Aires, IQUIBICEN, Ciudad de Buenos Aires, Argentina.,Laboratorio de Biología Molecular de Trypanosoma, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos, Ciudad de Buenos Aires, Argentina.,Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
8
|
Bonavita R, Laukkanen MO. Common Signal Transduction Molecules Activated by Bacterial Entry into a Host Cell and by Reactive Oxygen Species. Antioxid Redox Signal 2021; 34:486-503. [PMID: 32600071 DOI: 10.1089/ars.2019.7968] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Significance: An increasing number of pathogens are acquiring resistance to antibiotics. Efficient antimicrobial drug regimens are important even for the most advanced therapies, which range from cutting-edge invasive clinical protocols, such as robotic surgeries, to the treatment of harmless bacterial diseases and to minor scratches to the skin. Therefore, there is an urgent need to survey alternative antimicrobial drugs that can reinforce or replace existing antibiotics. Recent Advances: Bacterial proteins that are critical for energy metabolism, promising novel anticancer thiourea derivatives, and the use of synthetic molecules that increase the sensitivity of currently used antibiotics are among the recently discovered antimicrobial drugs. Critical Issues: In the development of new drugs, serious consideration should be given to the previous bacterial evolutionary selection caused by antibiotics, by the high proliferation rate of bacteria, and by the simple prokaryotic structure of bacteria. Future Directions: The survey of drug targets has mainly focused on bacterial proteins, although host signaling molecules involved in the treatment of various pathologies may have unknown antimicrobial characteristics. Recent data have suggested that small molecule inhibitors might enhance the effect of antibiotics, for example, by limiting bacterial entry into host cells. Phagocytosis, the mechanism by which host cells internalize pathogens through β-actin cytoskeletal rearrangement, induces calcium signaling, small GTPase activation, and phosphorylation of the phosphatidylinositol 3-kinase-serine/threonine-specific protein kinase B pathway. Antioxid. Redox Signal. 34, 486-503.
Collapse
Affiliation(s)
- Raffaella Bonavita
- Experimental Institute of Endocrinology and Oncology G. Salvatore, IEOS CNR, Naples, Italy
| | | |
Collapse
|
9
|
Fan S, Zhang D, Liu F, Yang Y, Xu H. Artesunate alleviates myocardial ischemia/reperfusion-induced myocardial necrosis in rats and hypoxia/reoxygenation-induced apoptosis in H9C2 cells via regulating the FAK/PI3K/Akt pathway. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1291. [PMID: 33209871 PMCID: PMC7661874 DOI: 10.21037/atm-20-5182] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background The various anti-inflammatory, anti-apoptotic, and antioxidant effects of Artesunate (Art) have been explored in numerous studies. This study aimed to evaluate the function of Art on myocardial necrosis in apoptotic cardiomyocytes in vivo and in vitro. Methods Sprague Dawley (SD) rats were randomly divided into groups: a control group, a myocardial ischemia reperfusion (MI/R) group, and MI/R+ Art groups. To establish a MI/R model, rats were subjected to left anterior descending artery ischemia for 45 minutes, and then reperfusion for 2 hours. Hypoxia was induced in H9C2 cells by subjecting them to hypoxic conditions at 37 °C for 4 hours, before placing them in a normoxic chamber for 2 hours. The test methods were used in this test, such as echocardiography, enzyme-linked immunosorbent assay (ELISA), HE staining, TUNEL staining, immunohistochemistry, flow cytometry, western blot, and CCK-8 assay. Results Art improved myocardial systolic function caused by MI/R injury in vivo. Simultaneously, Art reduced the levels of cardiac troponin I (cTnl), creatine kinase-MB (CK-MB) and myohemoglobin (Mb) in vivo and in vitro. Moreover, Art inhibited cardiomyocyte apoptosis in vivo and in vitro. The focal adhesion kinase (FAK)/phosphatidylinositide-3 kinases (PI3K)/AKT signaling pathway was also activated by Art in vivo and in vitro. Furthermore, after inhibitor PF573228 was added, Art inhibited apoptosis in H9C2 cells via activation of the FAK/PI3K/AKT signaling pathway in vitro. Conclusions This study confirms that Art alleviated MI/R injury and inhibited cardiomyocyte apoptosis in vivo and in vitro. Art exerted an inhibitory effect on cardiomyocyte apoptosis by activating the FAK/PI3K/AKT signaling pathway. Therefore, Art may serve as an alternative treatment for MI/R injury.
Collapse
Affiliation(s)
- Shunyang Fan
- Department of Heart Center, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Deyin Zhang
- Department of Galactphore, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fuyun Liu
- Department of Pediatric Orthopaedic, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuqi Yang
- Department of Heart Center, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongliang Xu
- Department of Heart Center, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
10
|
Melo TG, Coutinho EA, Pereira MCS. Heparan sulfate proteoglycan triggers focal adhesion kinase signaling during Trypanosoma cruzi invasion. Mem Inst Oswaldo Cruz 2020. [PMCID: PMC7849177 DOI: 10.1590/0074-02760200143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Trypanosoma cruzi, the etiologic agent of Chagas disease, is capable of triggering different signaling pathways that modulate its internalisation in mammalian cells. Focal adhesion kinase (FAK), a non-receptor tyrosine kinase protein, has been demonstrated as a mechanism of T. cruzi invasion in cardiomyocytes. Since the involved cell surface receptors are not yet known, we evaluated whether heparan sulfate proteoglycans (HSPG), a molecule involved in T. cruzi recognition and in the regulation of multiple signaling pathways, are able to trigger the FAK signaling pathway during T. cruzi invasion. METHODS To investigate the role of HSPG in the regulation of the FAK signaling pathway during trypomastigote entry, we performed heparan sulfate (HS) depletion from the cardiomyocyte surface by treatment with heparinase I or p-nitrophenyl-β-D-xylopyranoside (p-n-xyloside), which abolishes glycosaminoglycan (GAG) attachment to the proteoglycan core protein. Wild-type (CHO-k1) and GAG-deficient Chinese hamster ovary cells (CHO-745) were also used as an approach to evaluate the participation of the HSPG-FAK signaling pathway. FAK activation (FAK Tyr397) and spatial distribution were analysed by immunoblotting and indirect immunofluorescence, respectively. FINDINGS HS depletion from the cardiomyocyte surface inhibited FAK activation by T. cruzi. Cardiomyocyte treatment with heparinase I or p-n-xyloside resulted in 34% and 28% FAK phosphorylation level decreases, respectively. The experiments with the CHO cells corroborated the role of HSPG as a FAK activation mediator. T. cruzi infection did not stimulate FAK phosphorylation in CHO-745 cells, leading to a 36% reduction in parasite invasion. FAK inhibition due to the PF573228 treatment also impaired T. cruzi entry in CHO-k1 cells. MAIN CONCLUSION Jointly, our data demonstrate that HSPG is a key molecule in the FAK signaling pathway activation, regulating T. cruzi entry.
Collapse
|