1
|
Koh CC, Gollob KJ, Dutra WO. Cytokine Networks and the Clinical Outcome of American Teg-Umentary Leishmaniasis: Unveiling Targets for Alternative Therapeutic Interventions. Pathogens 2025; 14:188. [PMID: 40005563 PMCID: PMC11858318 DOI: 10.3390/pathogens14020188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 02/27/2025] Open
Abstract
American Tegumentary Leishmaniasis (ATL), caused by parasites of the genus Leishmania, presents a significant global health challenge, especially in Brazil, where cutaneous and mucosal forms are highly prevalent. Cutaneous Leishmaniasis (CL) typically results in single lesions, while mucosal Leishmaniasis (ML) leads to destructive mucosal lesions with a worse prognosis. The immune response, regulated by cytokines, plays a crucial role in disease progression and resolution. In CL, a balance between pro-inflammatory and anti-inflammatory cytokines is associated with lesion resolution, whereas in ML, an exaggerated inflammatory response worsens tissue damage. Thus, understanding cytokine regulation is essential for unveiling disease pathology and developing effective immunotherapeutic strategies. Here we discuss gene polymorphisms and epigenetic modifications that affect cytokine expression, influencing disease susceptibility and severity, as well as immunotherapeutic approaches that involve cytokine function in Leishmaniasis. In addition, we examine advancements in drug discovery, utilizing in silico methods and targeted drug delivery systems, providing potential avenues for better therapeutic interventions. Continuous research into immune responses and cytokine production and function is critical for identifying novel therapeutic targets and optimizing patient care for ATL.
Collapse
Affiliation(s)
- Carolina Cattoni Koh
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Av. Pres. Antônio Carlos, 6627-Pampulha, Belo Horizonte 31270-901, MG, Brazil
- National Institute of Science and Technology in Tropical Diseases, INCT-DT, Salvador 40110-160, BA, Brazil;
| | - Kenneth J. Gollob
- National Institute of Science and Technology in Tropical Diseases, INCT-DT, Salvador 40110-160, BA, Brazil;
- Hospital Israelita Albert Einstein, São Paulo 05652-900, SP, Brazil
| | - Walderez O. Dutra
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Av. Pres. Antônio Carlos, 6627-Pampulha, Belo Horizonte 31270-901, MG, Brazil
- National Institute of Science and Technology in Tropical Diseases, INCT-DT, Salvador 40110-160, BA, Brazil;
| |
Collapse
|
2
|
Nunes S, Bastos R, Marinho AI, Vieira R, Benício I, de Noronha MA, Lírio S, Brodskyn C, Tavares NM. Recent advances in the development and clinical application of miRNAs in infectious diseases. Noncoding RNA Res 2025; 10:41-54. [PMID: 39296638 PMCID: PMC11406675 DOI: 10.1016/j.ncrna.2024.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 08/06/2024] [Accepted: 09/01/2024] [Indexed: 09/21/2024] Open
Abstract
In the search for new biomarkers and therapeutic targets for infectious diseases, several molecules have been investigated. Small RNAs, known as microRNAs (miRs), are important regulators of gene expression, and have emerged as promising candidates for these purposes. MiRs are a class of small, endogenous non-coding RNAs that play critical roles in several human diseases, including host-pathogen interaction mechanisms. Recently, miRs signatures have been reported in different infectious diseases, opening new perspectives for molecular diagnosis and therapy. MiR profiles can discriminate between healthy individuals and patients, as well as distinguish different disease stages. Furthermore, the possibility of assessing miRs in biological fluids, such as serum and whole blood, renders these molecules feasible for the development of new non-invasive diagnostic and prognostic tools. In this manuscript, we will comprehensively describe miRs as biomarkers and therapeutic targets in infectious diseases and explore how they can contribute to the advance of existing and new tools. Additionally, we will discuss different miR analysis platforms to understand the obstacles and advances of this molecular approach and propose their potential clinical applications and contributions to public health.
Collapse
Affiliation(s)
- Sara Nunes
- Laboratory of Medicine and Precision Public Health (MeSP), Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Bahia, Brazil
| | - Rana Bastos
- Laboratory of Medicine and Precision Public Health (MeSP), Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Bahia, Brazil
- Federal University of Bahia (UFBA), Salvador, Brazil
| | - Ananda Isis Marinho
- Laboratory of Medicine and Precision Public Health (MeSP), Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Bahia, Brazil
- Federal University of Bahia (UFBA), Salvador, Brazil
| | - Raissa Vieira
- Laboratory of Medicine and Precision Public Health (MeSP), Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Bahia, Brazil
- Federal University of Bahia (UFBA), Salvador, Brazil
| | - Ingra Benício
- Laboratory of Medicine and Precision Public Health (MeSP), Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Bahia, Brazil
| | | | - Sofia Lírio
- Laboratory of Medicine and Precision Public Health (MeSP), Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Bahia, Brazil
- Bahiana School of Medicine and Public Health, Salvador, Brazil
| | - Cláudia Brodskyn
- Federal University of Bahia (UFBA), Salvador, Brazil
- Laboratory of Parasite-Host Interaction and Epidemiology (LaIPHE), Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Bahia, Brazil
- Instituto Nacional de Ciência e Tecnologia (INCT) Iii - Instituto de Investigação Em Imunologia, São Paulo, Brazil
| | - Natalia Machado Tavares
- Laboratory of Medicine and Precision Public Health (MeSP), Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Bahia, Brazil
- Federal University of Bahia (UFBA), Salvador, Brazil
- Instituto Nacional de Ciência e Tecnologia (INCT) Iii - Instituto de Investigação Em Imunologia, São Paulo, Brazil
| |
Collapse
|
3
|
Song Y, Qiu J, Kang J, Chen Y, Cao R, Wang W, Dai M, Chen D, Fu Z, Guo R. Transcriptomic Characterization of miRNAs in Apis cerana Larvae Responding to Ascosphaera apis Infection. Genes (Basel) 2025; 16:156. [PMID: 40004485 PMCID: PMC11855511 DOI: 10.3390/genes16020156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 02/27/2025] Open
Abstract
Ascosphaera apis is a fungal pathogen that specifically infects bee larvae, causing an outbreak of chalkbrood disease in the bee colony and a decline in the number of bee colonies. The role of miRNA regulation in honeybees in response to A. apis infection is unclear. In this study, based on small RNA-seq, we identified the differentially expressed miRNAs (DEmiRNAs) and their regulatory networks and functions in the gut of Apis cerana cerana on the first day (AcT1), the second day (AcT2) and the third day (AcT3) after A. apis infection, and analyzed the immune response mechanism of A. apis through the miRNAs-mRNA regulation network of A. apis infection. A total of 537 miRNAs were obtained, and 10, 27, and 54 DEmiRNAs were screened in the AcT1, AcT2, and AcT3 groups, respectively. The number of DEmiRNAs gradually increased with the infection time. Stem-loop RT-PCR results showed that most of the DEmiRNAs were truly expressed, and the expression trend of DEmiRNAs was consistent with the results of sRNA-seq. The top five GO terms of DEmiRNA-targeted mRNA were binding, cellular process, catalytic activity, metabolic process, and single-organism process. The main pathways enriched by KEGG were endocytosis, ubiquitin-mediated proteolysis, phagosome, and the JAK-STAT immune-related signaling pathways. The number of DEmiRNAs and target mRNAs of these related pathway genes increased with infection time. The miRNA-mRNA regulatory network analysis showed that ace-miR-539-y was the core miRNA of the early immune response in the gut of larvae infected with A. apis in the JAK-STAT pathway and phagosome, and ace-miR-1277-x was the core miRNA of the late immune response in the gut of larvae infected with A. apis in the JAK-STAT signaling pathway and phagosome. The results showed that miRNA participated in the immune response of honeybees to A. apis infection by regulating the host's energy metabolism, cellular immunity, and humoral immunity. The results of this study provide a basis for the regulation of miRNAs in A. c. cerana larvae in response to A. apis infection and provide new insights into host-pathogen interactions.
Collapse
Affiliation(s)
- Yuxuan Song
- College of Bee Science and Biomedicine, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (Y.S.); (J.Q.); (J.K.); (Y.C.); (R.C.); (W.W.); (M.D.); (D.C.); (Z.F.)
| | - Jianfeng Qiu
- College of Bee Science and Biomedicine, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (Y.S.); (J.Q.); (J.K.); (Y.C.); (R.C.); (W.W.); (M.D.); (D.C.); (Z.F.)
- National & Local United Engineering Laboratory of Natural Biotoxin, Fuzhou 350002, China
- Apitherapy Research Institute, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Jing Kang
- College of Bee Science and Biomedicine, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (Y.S.); (J.Q.); (J.K.); (Y.C.); (R.C.); (W.W.); (M.D.); (D.C.); (Z.F.)
| | - Ying Chen
- College of Bee Science and Biomedicine, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (Y.S.); (J.Q.); (J.K.); (Y.C.); (R.C.); (W.W.); (M.D.); (D.C.); (Z.F.)
| | - Ruihua Cao
- College of Bee Science and Biomedicine, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (Y.S.); (J.Q.); (J.K.); (Y.C.); (R.C.); (W.W.); (M.D.); (D.C.); (Z.F.)
| | - Wei Wang
- College of Bee Science and Biomedicine, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (Y.S.); (J.Q.); (J.K.); (Y.C.); (R.C.); (W.W.); (M.D.); (D.C.); (Z.F.)
| | - Mengyuan Dai
- College of Bee Science and Biomedicine, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (Y.S.); (J.Q.); (J.K.); (Y.C.); (R.C.); (W.W.); (M.D.); (D.C.); (Z.F.)
| | - Dafu Chen
- College of Bee Science and Biomedicine, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (Y.S.); (J.Q.); (J.K.); (Y.C.); (R.C.); (W.W.); (M.D.); (D.C.); (Z.F.)
- National & Local United Engineering Laboratory of Natural Biotoxin, Fuzhou 350002, China
- Apitherapy Research Institute, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Zhongmin Fu
- College of Bee Science and Biomedicine, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (Y.S.); (J.Q.); (J.K.); (Y.C.); (R.C.); (W.W.); (M.D.); (D.C.); (Z.F.)
- National & Local United Engineering Laboratory of Natural Biotoxin, Fuzhou 350002, China
- Apitherapy Research Institute, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Rui Guo
- College of Bee Science and Biomedicine, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (Y.S.); (J.Q.); (J.K.); (Y.C.); (R.C.); (W.W.); (M.D.); (D.C.); (Z.F.)
- National & Local United Engineering Laboratory of Natural Biotoxin, Fuzhou 350002, China
- Apitherapy Research Institute, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| |
Collapse
|
4
|
Huang Z, Cao M, Fan H, Sun Y, Lan T, Ma J, Li Q. Porcine delta coronavirus inhibits NHE3 activity of porcine intestinal epithelial cells through miR-361-3p/NHE3 regulatory axis. Vet Microbiol 2024; 289:109916. [PMID: 38159369 DOI: 10.1016/j.vetmic.2023.109916] [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: 08/02/2023] [Revised: 11/05/2023] [Accepted: 11/17/2023] [Indexed: 01/03/2024]
Abstract
Porcine deltacoronavirus (PDCoV) infection in piglets can cause small intestinal epithelial necrosis and atrophic enteritis, which leads to severe damages to host cells, and result in diarrhea. In this study, we investigated the relationship between miR-361, SLC9A3(Solute carrier family 9, subfamily A, member 3), and NHE3(sodium-hydrogen exchanger member 3) in in porcine intestinal epithelial cells (IPI-2I) cells after PDCoV infection. Our results showed that the ssc-miR-361-3p expression inhibits the mRNA level of SLC9A3 gene which lead to the descending of NHE3 protein expression, and the NHE3 activity was suppressed. NHE3 activity was suppressed via down-regulation expression of SLC9A3 mRNA by transfection with siRNA. Ssc-miR-361-3p mimics and inhibitors were used to change the expression of ssc-miR-361-3p in IPI-2I cells. Ssc-miR-361-3p overexpression reduced the mRNA level of SLC9A3 gene, the level of NHE3 protein expression and NHE3 activity in IPI-2I cells, while ssc-miR-361-3p inhibits NHE3. Furthermore, luciferase reporter assay showed that SLC9A3 gene was a direct target of ssc-miR-361-3p. Ssc-miR-361-3p inhibition restored NHE3 activity in PDCoV infected IPI-2I cells by up-regulating SLC9A3 mRNA expression and NHE3 protein expression. These results demonstrate that the PDCoV infection can inhibit NHE3 activity through miR-361-3p/SLC9A3 regulatory axis. The relevant research is reported for the first time in PDCoV, which has significance in exploring the pathogenic mechanism of PDCoV and can provide a theoretical basis for its prevention and control. suggesting that NHE3 and ssc-miR-361-3p may be potential therapeutic targets for diarrhea in infected piglets.
Collapse
Affiliation(s)
- Zehong Huang
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510642, China
| | - Mengjing Cao
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510642, China
| | - Haoqian Fan
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510642, China
| | - Yuan Sun
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510642, China
| | - Tian Lan
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510642, China
| | - Jingyun Ma
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510642, China.
| | - Qianniu Li
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510642, China.
| |
Collapse
|
5
|
Lago T, Medina L, Lago J, Santana N, Cardoso T, Rocha A, Leal-Calvo T, Carvalho EM, Castellucci LC. MicroRNAs regulating macrophages infected with Leishmania L. ( V.) Braziliensis isolated from different clinical forms of American tegumentary leishmaniasis. Front Immunol 2023; 14:1280949. [PMID: 38143766 PMCID: PMC10748487 DOI: 10.3389/fimmu.2023.1280949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 11/20/2023] [Indexed: 12/26/2023] Open
Abstract
Background Leishmaniasis is an infectious disease caused by protozoa of the genus Leishmania. There are still no vaccines, and therapeutic options are limited, indicating the constant need to understand the fine mechanisms of its pathophysiology. An approach that has been explored in leishmaniasis is the participation of microRNAs (miRNAs), a class of small non-coding RNAs that act, in most cases, to repress gene expression. miRNAs play a role in the complex and plastic interaction between the host and pathogens, either as part of the host's immune response to neutralize infection or as a molecular strategy employed by the pathogen to modulate host pathways to its own benefit. Methods Monocyte-derived macrophages from healthy subjects were infected with isolates of three clinical forms of L. braziliensis: cutaneous (CL), mucosal (ML), and disseminated (DL) leishmaniasis. We compared the expression of miRNAs that take part in the TLR/NFkB pathways. Correlations with parasite load as well as immune parameters were analyzed. Results miRNAs -103a-3p, -21-3p, 125a-3p -155-5p, -146a-5p, -132- 5p, and -147a were differentially expressed in the metastatic ML and DL forms, and there was a direct correlation between miRNAs -103a-3p, -21-3p, -155-5p, -146a-5p, -132-5p, and -9-3p and parasite load with ML and DL isolates. We also found a correlation between the expression of miR-21-3p and miR-146a-5p with the antiapoptotic gene BCL2 and the increase of viable cells, whereas miR-147a was indirectly correlated with CXCL-9 levels. Conclusion The expression of miRNAs is strongly correlated with the parasite load and the inflammatory response, suggesting the participation of these molecules in the pathogenesis of the different clinical forms of L. braziliensis.
Collapse
Affiliation(s)
- Tainã Lago
- Serviço de Imunologia da Universidade Federal da Bahia, Salvador, Brazil
- Programa de Pós-graduação em Ciências da Saúde da Universidade Federal da Bahia, Salvador, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), Ministério da Ciência, Tecnologia, Inovações e Comunicações, CNPq, Brasília, DF, Brazil
| | - Lilian Medina
- Serviço de Imunologia da Universidade Federal da Bahia, Salvador, Brazil
- Programa de Pós-graduação em Ciências da Saúde da Universidade Federal da Bahia, Salvador, Brazil
| | - Jamile Lago
- Serviço de Imunologia da Universidade Federal da Bahia, Salvador, Brazil
- Programa de Pós-graduação em Ciências da Saúde da Universidade Federal da Bahia, Salvador, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), Ministério da Ciência, Tecnologia, Inovações e Comunicações, CNPq, Brasília, DF, Brazil
| | - Nadja Santana
- Serviço de Imunologia da Universidade Federal da Bahia, Salvador, Brazil
- Programa de Pós-graduação em Ciências da Saúde da Universidade Federal da Bahia, Salvador, Brazil
| | - Thiago Cardoso
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), Ministério da Ciência, Tecnologia, Inovações e Comunicações, CNPq, Brasília, DF, Brazil
- Laboratório de Pesquisas Clínicas (LAPEC), Instituto Gonçalo Moniz-FIOCRUZ, Sakvador, Bahia, Brazil
| | - Alan Rocha
- Laboratório de Pesquisas Clínicas (LAPEC), Instituto Gonçalo Moniz-FIOCRUZ, Sakvador, Bahia, Brazil
| | | | - Edgar M. Carvalho
- Serviço de Imunologia da Universidade Federal da Bahia, Salvador, Brazil
- Programa de Pós-graduação em Ciências da Saúde da Universidade Federal da Bahia, Salvador, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), Ministério da Ciência, Tecnologia, Inovações e Comunicações, CNPq, Brasília, DF, Brazil
- Laboratório de Pesquisas Clínicas (LAPEC), Instituto Gonçalo Moniz-FIOCRUZ, Sakvador, Bahia, Brazil
| | - Léa Cristina Castellucci
- Serviço de Imunologia da Universidade Federal da Bahia, Salvador, Brazil
- Programa de Pós-graduação em Ciências da Saúde da Universidade Federal da Bahia, Salvador, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), Ministério da Ciência, Tecnologia, Inovações e Comunicações, CNPq, Brasília, DF, Brazil
| |
Collapse
|
6
|
Naouar I, Kammoun Rebai W, Ben Salah A, Bouguerra H, Toumi A, Hamida NB, Louzir H, Meddeb-Garnaoui A. A Prospective cohort study of zoonotic cutaneous leishmaniasis in tunisia: Clinical and Immunological features and immune correlates of protection. PLoS Negl Trop Dis 2023; 17:e0011784. [PMID: 38064516 PMCID: PMC10732404 DOI: 10.1371/journal.pntd.0011784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 12/20/2023] [Accepted: 11/09/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND This study aimed to define immunological markers of exposure to L. major parasites and identify correlates of protection against infection. METHODS We analyzed a cohort of 790 individuals at risk of developing ZCL living in endemic areas with varying L. major infection prevalence. One area had a high infection prevalence indicated by high proportions of leishmanin skin test (LST) positive subjects, while the other areas were recent foci with lower infection prevalence. Blood samples were collected before the transmission season to measure Interferon gamma (IFN-γ), Interleukin 10 (IL-10), and Granzyme B (GrB) levels in response to parasite stimulation in peripheral blood mononuclear cells. A one-year follow-up period involved active detection of new ZCL cases to estimate disease incidence after a transmission season and identify immune correlates of protection. RESULTS The study population showed heterogeneity in parasite contact, evident from specific scars and/or positive LST results, significantly higher in the old focus compared to recent foci. IFN-γ and GrB were markers of parasite exposure and reliable indicators of immunity to L. major. Positive correlations were observed between IFN-γ/IL-10 and GrB/IL-10 ratios and LST results. Unexpectedly, only 29 new ZCL cases (4%) appeared after a transmission season, with 27 cases reported in recent foci and 2 in the oldest focus. Our findings indicate that individuals in L. major endemic areas are likely to develop ZCL regardless of their LST status. We showed that high pre-transmission season levels of IFN-γ and GrB produced by PBMC, along with a high IFN-γ/IL-10 ratio, were associated with protection. CONCLUSION This study on a large cohort at risk of ZCL confirmed IFN-γ and GrB as protective factors against the disease. A high IFN-γ/IL-10 ratio, but not GrB/IL-10 ratio was associated with resistance. These results are valuable for developing and evaluating of a vaccine against human leishmaniasis.
Collapse
Affiliation(s)
- Ikbel Naouar
- Laboratory of Transmission Control and Immunobiology of Infection, Pasteur Institute of Tunis, Tunis, Tunisia
- Faculty of Sciences, University of Tunis El Manar, Tunis, Tunisia
- Department of Immunology, University of Toronto, Ontario, Canada
| | - Wafa Kammoun Rebai
- Faculty of Sciences, University of Tunis El Manar, Tunis, Tunisia
- Laboratory of Medical Parasitology, Biotechnology and Biomolecular, Pasteur Institute of Tunis, Tunis, Tunisia
| | - Afif Ben Salah
- Laboratory of Transmission Control and Immunobiology of Infection, Pasteur Institute of Tunis, Tunis, Tunisia
- Department of Family and Community Medicine, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
| | - Hind Bouguerra
- National Observatory of New and Emerging Diseases, Tunis, Tunisia
- Faculty of Medicine of Tunis, University Tunis El Manar, Tunis, Tunisia
| | - Amine Toumi
- Health Information and Intelligence Section, Ministry of Public Health, Doha, Qatar
| | - Nabil Belhadj Hamida
- Laboratory of Transmission Control and Immunobiology of Infection, Pasteur Institute of Tunis, Tunis, Tunisia
| | - Hechmi Louzir
- Laboratory of Transmission Control and Immunobiology of Infection, Pasteur Institute of Tunis, Tunis, Tunisia
- National Observatory of New and Emerging Diseases, Tunis, Tunisia
| | - Amel Meddeb-Garnaoui
- Laboratory of Medical Parasitology, Biotechnology and Biomolecular, Pasteur Institute of Tunis, Tunis, Tunisia
| |
Collapse
|
7
|
Soares MF, Costa SF, de Freitas JH, Rebech GT, Dos Santos MO, de Lima VMF. MiR-150 regulates the Leishmania infantum parasitic load and granzyme B levels in peripheral blood mononuclear cells of dogs with canine visceral leishmaniosis. Vet Parasitol 2023; 320:109958. [PMID: 37269731 DOI: 10.1016/j.vetpar.2023.109958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/17/2023] [Accepted: 05/20/2023] [Indexed: 06/05/2023]
Abstract
Leishmania infantum causes visceral leishmaniosis, a neglected tropical disease that can modulate the host immune response by altering the expression of small non-coding RNAs called microRNAs (miRNAs). Some miRNAs are differentially expressed in peripheral blood mononuclear cells (PBMCs) of dogs with canine visceral leishmaniosis (CanL), like the down-regulated miR-150. Even though miR-150 is negatively correlated with L. infantum parasitic load, it is unclear if miR-150 directly affects L. infantum parasitic load and (if so) how this miRNA would contribute to infection. Here, we isolated PBMCs from 14 naturally infected dogs (CanL group) and six healthy dogs (Control group) and treated them in vitro with miR-150 mimic or inhibitor. We measured L. infantum parasitic load using qPCR and compared treatments. We also measured miR-150 in silico predicted target protein levels (STAT1, TNF-α, HDAC8, and GZMB) using flow cytometry or enzyme-linked immunosorbent assays. Increasing miR-150 activity diminished L. infantum parasitic load in CanL PBMCs. We also found that inhibition of miR-150 reduced GZMB (granzyme B) levels. These findings demonstrate that miR-150 plays an important role in L. infantum infection in canine PBMCs, and they merit further studies aiming at drug development.
Collapse
Affiliation(s)
- Matheus Fujimura Soares
- Department of Clinical Medicine, Surgery and Animal Reproduction, São Paulo State University (UNESP), School of Veterinary Medicine, 793 Clóvis Pestana St., Araçatuba, São Paulo, 16050-680, Brazil
| | - Sidnei Ferro Costa
- Department of Clinical Medicine, Surgery and Animal Reproduction, São Paulo State University (UNESP), School of Veterinary Medicine, 793 Clóvis Pestana St., Araçatuba, São Paulo, 16050-680, Brazil
| | - Jéssica Henrique de Freitas
- Department of Clinical Medicine, Surgery and Animal Reproduction, São Paulo State University (UNESP), School of Veterinary Medicine, 793 Clóvis Pestana St., Araçatuba, São Paulo, 16050-680, Brazil
| | - Gabriela Torres Rebech
- Department of Clinical Medicine, Surgery and Animal Reproduction, São Paulo State University (UNESP), School of Veterinary Medicine, 793 Clóvis Pestana St., Araçatuba, São Paulo, 16050-680, Brazil
| | - Marilene Oliveira Dos Santos
- Department of Clinical Medicine, Surgery and Animal Reproduction, São Paulo State University (UNESP), School of Veterinary Medicine, 793 Clóvis Pestana St., Araçatuba, São Paulo, 16050-680, Brazil
| | - Valéria Marçal Felix de Lima
- Department of Clinical Medicine, Surgery and Animal Reproduction, São Paulo State University (UNESP), School of Veterinary Medicine, 793 Clóvis Pestana St., Araçatuba, São Paulo, 16050-680, Brazil.
| |
Collapse
|
8
|
Zou X, Liu T, Huang Z, Zhou W, Yuan M, Zhao H, Pan Z, Chen P, Shao Y, Hu X, Zhang S, Zheng S, Zhang Y, Huang P. SOX17 is a Critical Factor in Maintaining Endothelial Function in Pulmonary Hypertension by an Exosome-Mediated Autocrine Manner. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206139. [PMID: 36919784 PMCID: PMC10190640 DOI: 10.1002/advs.202206139] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 02/20/2023] [Indexed: 05/18/2023]
Abstract
Endothelial dysfunction is considered a predominant driver for pulmonary vascular remodeling in pulmonary hypertension (PH). SOX17, a key regulator of vascular homoeostasis, has been found to harbor mutations in PH patients, which are associated with PH susceptibility. Here, this study explores whether SOX17 mediates the autocrine activity of pulmonary artery ECs to maintain endothelial function and vascular homeostasis in PH and its underlying mechanism. It is found that SOX17 expression is downregulated in the endothelium of remodeled pulmonary arteries in IPH patients and SU5416/hypoxia (Su/hypo)-induced PH mice as well as dysfunctional HPAECs. Endothelial knockdown of SOX17 accelerates the progression of Su/hypo-induced PH in mice. SOX17 overexpression in the pulmonary endothelium of mice attenuates Su/hypo-induced PH. SOX17-associated exosomes block the proliferation, apoptosis, and inflammation of HPAECs, preventing pulmonary arterial remodeling and Su/hypo-induced PH. Mechanistic analyses demonstrates that overexpressing SOX17 promotes the exosome-mediated release of miR-224-5p and miR-361-3p, which are internalized by injured HPAECs in an autocrine manner, ultimately repressing the upregulation of NR4A3 and PCSK9 genes and improving endothelial function. These results suggest that SOX17 is a key gene in maintaining endothelial function and vascular homeostasis in PH through regulating exosomal miRNAs in an autocrine manner.
Collapse
Affiliation(s)
- Xiaozhou Zou
- Center for Clinical PharmacyCancer CenterDepartment of PharmacyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhou310014P. R. China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceHangzhou310014P. R. China
| | - Ting Liu
- Department of PharmacyAffiliated Hangzhou First People's HospitalZhejiang University School of MedicineHangzhou310006P. R. China
- Department of Clinical PharmacyKey Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang ProvinceAffiliated Hangzhou First People's HospitalZhejiang University School of MedicineHangzhou310006P. R. China
| | - Zhongjie Huang
- School of Pharmaceutical SciencesZhejiang Chinese Medical UniversityHangzhou310014P. R. China
| | - Wei Zhou
- Zhongnan Hospital of Wuhan UniversityInstitute of Hepatobiliary Diseases of Wuhan UniversityTransplant Center of Wuhan UniversityHubei Key Laboratory of Medical Technology on TransplantationWuhan430000P. R. China
| | - Mengnan Yuan
- Center for Clinical PharmacyCancer CenterDepartment of PharmacyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhou310014P. R. China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceHangzhou310014P. R. China
| | - Hongying Zhao
- Center for Clinical PharmacyCancer CenterDepartment of PharmacyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhou310014P. R. China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceHangzhou310014P. R. China
| | - Zongfu Pan
- Center for Clinical PharmacyCancer CenterDepartment of PharmacyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhou310014P. R. China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceHangzhou310014P. R. China
| | - Pengcheng Chen
- Center for Clinical PharmacyCancer CenterDepartment of PharmacyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhou310014P. R. China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceHangzhou310014P. R. China
| | - Yanfei Shao
- Center for Clinical PharmacyCancer CenterDepartment of PharmacyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhou310014P. R. China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceHangzhou310014P. R. China
| | - Xiaoping Hu
- Center for Clinical PharmacyCancer CenterDepartment of PharmacyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhou310014P. R. China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceHangzhou310014P. R. China
| | - Su Zhang
- Center for Clinical PharmacyCancer CenterDepartment of PharmacyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhou310014P. R. China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceHangzhou310014P. R. China
| | - Shuilian Zheng
- Center for Clinical PharmacyCancer CenterDepartment of PharmacyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhou310014P. R. China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceHangzhou310014P. R. China
| | - Yiwen Zhang
- Center for Clinical PharmacyCancer CenterDepartment of PharmacyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhou310014P. R. China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceHangzhou310014P. R. China
| | - Ping Huang
- Center for Clinical PharmacyCancer CenterDepartment of PharmacyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhou310014P. R. China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceHangzhou310014P. R. China
| |
Collapse
|
9
|
Mahor H, Mukherjee A, Sarkar A, Saha B. Anti-leishmanial therapy: Caught between drugs and immune targets. Exp Parasitol 2023; 245:108441. [PMID: 36572088 DOI: 10.1016/j.exppara.2022.108441] [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: 05/11/2022] [Revised: 08/12/2022] [Accepted: 12/01/2022] [Indexed: 12/24/2022]
Abstract
Leishmaniasis is an enigmatic disease that has very restricted options for chemotherapy and none for prophylaxis. As a result, deriving therapeutic principles for curing the disease has been a major objective in Leishmania research for a long time. Leishmania is a protozoan parasite that lives within macrophages by subverting or switching cell signaling to the pathways that ensure its intracellular survival. Therefore, three groups of molecules aimed at blocking or eliminating the parasite, at least, in principle, include blockers of macrophage receptor- Leishmania ligand interaction, macrophage-activating small molecules, peptides and cytokines, and signaling inhibitors or activators. Macrophages also act as an antigen-presenting cell, presenting antigen to the antigen-specific T cells to induce activation and differentiation of the effector T cell subsets that either execute or suppress anti-leishmanial functions. Three groups of therapeutic principles targeting this sphere of Leishmania-macrophage interaction include antibodies that block pro-leishmanial response of T cells, ligands that activate anti-leishmanial T cells and the antigens for therapeutic vaccines. Besides these, prophylactic vaccines have been in clinical trials but none has succeeded so far. Herein, we have attempted to encompass all these principles and compose a comprehensive review to analyze the feasibility and adoptability of different therapeutics for leishmaniasis.
Collapse
Affiliation(s)
- Hima Mahor
- National Centre for Cell Science, Ganeshkhind, Pune, 411007, India
| | - Arka Mukherjee
- Trident Academy of Creative Technology, Bhubaneswar, 751024, Odisha, India
| | - Arup Sarkar
- Trident Academy of Creative Technology, Bhubaneswar, 751024, Odisha, India
| | - Bhaskar Saha
- National Centre for Cell Science, Ganeshkhind, Pune, 411007, India; Trident Academy of Creative Technology, Bhubaneswar, 751024, Odisha, India.
| |
Collapse
|
10
|
Gupta AK, Das S, Kamran M, Ejazi SA, Ali N. The Pathogenicity and Virulence of Leishmania - interplay of virulence factors with host defenses. Virulence 2022; 13:903-935. [PMID: 35531875 PMCID: PMC9154802 DOI: 10.1080/21505594.2022.2074130] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Leishmaniasis is a group of disease caused by the intracellular protozoan parasite of the genus Leishmania. Infection by different species of Leishmania results in various host immune responses, which usually lead to parasite clearance and may also contribute to pathogenesis and, hence, increasing the complexity of the disease. Interestingly, the parasite tends to reside within the unfriendly environment of the macrophages and has evolved various survival strategies to evade or modulate host immune defense. This can be attributed to the array of virulence factors of the vicious parasite, which target important host functioning and machineries. This review encompasses a holistic overview of leishmanial virulence factors, their role in assisting parasite-mediated evasion of host defense weaponries, and modulating epigenetic landscapes of host immune regulatory genes. Furthermore, the review also discusses the diagnostic potential of various leishmanial virulence factors and the advent of immunomodulators as futuristic antileishmanial drug therapy.
Collapse
Affiliation(s)
- Anand Kumar Gupta
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Sonali Das
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Mohd Kamran
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Sarfaraz Ahmad Ejazi
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Nahid Ali
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| |
Collapse
|
11
|
Ozturk EA, Caner A. Liquid Biopsy for Promising Non-invasive Diagnostic Biomarkers in Parasitic Infections. Acta Parasitol 2022; 67:1-17. [PMID: 34176040 DOI: 10.1007/s11686-021-00444-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 06/16/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Liquid biopsy refers to the sampling and molecular analysis of body fluids such as blood, saliva, and urine in contrast to conventional tissue biopsies. Liquid biopsy approach can offer powerful non-invasive biomarkers (circulating markers) for diagnosis and monitoring treatment response of a variety of diseases, including parasitic infections. METHODS In this review, we concentrate on cell-free DNA (cfDNA), microRNA (miRNA), and exosomes in the published literature. RESULTS Considering the high prevalence and severity of parasitic infections worldwide, circulating biomarkers can provide a new insight into the diagnosis and prognosis of parasites in the near future. Moreover, identifying and characterizing parasite- or host-derived circulating markers are important for a better understanding of the pathogenesis of parasite infection and host-parasite relationship at the molecular level. Profiling of biomarkers for parasitic diseases is a promising potential field, though further studies and optimization strategies are required, both in vitro and in vivo. CONCLUSION In this review, we discuss three approaches in the liquid biopsy including circulating cfDNA, miRNAs, and exosomes for diagnosis and evaluation of parasites and summarize circulating biomarkers in non-invasive samples during parasitic infections.
Collapse
Affiliation(s)
- Eylem Akdur Ozturk
- Department of Parasitology, Çukurova University Faculty of Medicine, Adana, Turkey
| | - Ayse Caner
- Department of Parasitology, Ege University Faculty of Medicine, 35100, Izmir, Turkey.
- Cancer Research Center, Ege University, Izmir, Turkey.
| |
Collapse
|
12
|
MiR-361-3p alleviates cerebral ischemia–reperfusion injury by targeting NACC1 through the PINK1/Parkin pathway. J Mol Histol 2022; 53:357-367. [DOI: 10.1007/s10735-021-10049-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 12/07/2021] [Indexed: 10/19/2022]
|
13
|
Tamgue O, Mezajou CF, Ngongang NN, Kameni C, Ngum JA, Simo USF, Tatang FJ, Akami M, Ngono AN. Non-Coding RNAs in the Etiology and Control of Major and Neglected Human Tropical Diseases. Front Immunol 2021; 12:703936. [PMID: 34737736 PMCID: PMC8560798 DOI: 10.3389/fimmu.2021.703936] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 09/09/2021] [Indexed: 12/19/2022] Open
Abstract
Non-coding RNAs (ncRNAs) including microRNAs (miRs) and long non-coding RNAs (lncRNAs) have emerged as key regulators of gene expression in immune cells development and function. Their expression is altered in different physiological and disease conditions, hence making them attractive targets for the understanding of disease etiology and the development of adjunctive control strategies, especially within the current context of mitigated success of control measures deployed to eradicate these diseases. In this review, we summarize our current understanding of the role of ncRNAs in the etiology and control of major human tropical diseases including tuberculosis, HIV/AIDS and malaria, as well as neglected tropical diseases including leishmaniasis, African trypanosomiasis and leprosy. We highlight that several ncRNAs are involved at different stages of development of these diseases, for example miR-26-5p, miR-132-3p, miR-155-5p, miR-29-3p, miR-21-5p, miR-27b-3p, miR-99b-5p, miR-125-5p, miR-146a-5p, miR-223-3p, miR-20b-5p, miR-142-3p, miR-27a-5p, miR-144-5p, miR-889-5p and miR-582-5p in tuberculosis; miR-873, MALAT1, HEAL, LINC01426, LINC00173, NEAT1, NRON, GAS5 and lincRNA-p21 in HIV/AIDS; miR-451a, miR-let-7b and miR-106b in malaria; miR-210, miR-30A-5P, miR-294, miR-721 and lncRNA 7SL RNA in leishmaniasis; and miR-21, miR-181a, miR-146a in leprosy. We further report that several ncRNAs were investigated as diseases biomarkers and a number of them showed good potential for disease diagnosis, including miR-769-5p, miR-320a, miR-22-3p, miR-423-5p, miR-17-5p, miR-20b-5p and lncRNA LOC152742 in tuberculosis; miR-146b-5p, miR-223, miR-150, miR-16, miR-191 and lncRNA NEAT1 in HIV/AIDS; miR-451 and miR-16 in malaria; miR-361-3p, miR-193b, miR-671, lncRNA 7SL in leishmaniasis; miR-101, miR-196b, miR-27b and miR-29c in leprosy. Furthermore, some ncRNAs have emerged as potential therapeutic targets, some of which include lncRNAs NEAT1, NEAT2 and lnr6RNA, 152742 in tuberculosis; MALAT1, HEAL, SAF, lincRNA-p21, NEAT1, GAS5, NRON, LINC00173 in HIV/AIDS; miRNA-146a in malaria. Finally, miR-135 and miR-126 were proposed as potential targets for the development of therapeutic vaccine against leishmaniasis. We also identify and discuss knowledge gaps that warrant for increased research work. These include investigation of the role of ncRNAs in the etiology of African trypanosomiasis and the assessment of the diagnostic potential of ncRNAs for malaria, and African trypanosomiasis. The potential targeting of ncRNAs for adjunctive therapy against tuberculosis, leishmaniasis, African trypanosomiasis and leprosy, as well as their targeting in vaccine development against tuberculosis, HIV/AIDS, malaria, African trypanosomiasis and leprosy are also new avenues to explore.
Collapse
Affiliation(s)
- Ousman Tamgue
- Department of Biochemistry, Faculty of Sciences, University of Douala, Douala, Cameroon
| | | | | | - Charleine Kameni
- Department of Biochemistry, Faculty of Sciences, University of Douala, Douala, Cameroon
| | - Jubilate Afuoti Ngum
- Department of Biochemistry, Faculty of Sciences, University of Douala, Douala, Cameroon
| | | | - Fabrice Junior Tatang
- Department of Biochemistry, Faculty of Sciences, University of Douala, Douala, Cameroon
| | - Mazarin Akami
- Department of Biochemistry, Faculty of Sciences, University of Douala, Douala, Cameroon
| | - Annie Ngane Ngono
- Department of Biochemistry, Faculty of Sciences, University of Douala, Douala, Cameroon
| |
Collapse
|
14
|
Wang J, Xie Z, Liu Y, Zhang W, Ji T. MicroRNA-361 reduces the viability and migratory ability of pancreatic cancer cells via mediation of the MAPK/JNK pathway. Exp Ther Med 2021; 22:1365. [PMID: 34659511 PMCID: PMC8515516 DOI: 10.3892/etm.2021.10799] [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: 11/26/2019] [Accepted: 06/19/2020] [Indexed: 12/16/2022] Open
Abstract
Previous research has revealed that microRNA-361 (miR-361) functions as a fundamental modulator in non-small-cell lung cancer and esophageal carcinoma. However, its involvement in pancreatic cancer (PC) is yet to be elucidated. Therefore, the present study aimed to examine the mechanism and function of miR-361 during the regulation of PC cell migration and viability. It was demonstrated that miR-361 expression decreased in PC cell lines and tissues, and the overexpression of miR-361 suppressed in vivo PC cell proliferation in mice. Moreover, flow cytometry and MTT assays indicated that the miR-361 mimic decreased the viability and increased the apoptosis of PC cells. Both Transwell migration and wound healing assays identified that miR-361 ameliorated the migratory ability of PC cells. Using dual-luciferase reporter assays, it was found that miR-361 targeted mitogen-activated protein kinase (MAPK)/JNK 3'-untranslated regions, inducing the downregulation of this gene. In PC cells, overexpression of MAPK/JNK diminished the pro-apoptotic effect of the miR-361 mimic, while restoring the migratory activity of PC cells. Collectively, the present results suggested novel molecular mechanisms underlying PC progression and development.
Collapse
Affiliation(s)
- Juan Wang
- Department of Gastroenterology, People's Hospital of Leling City, Dezhou, Shandong 253600, P.R. China
| | - Zongjing Xie
- Department of General Surgery, Zhucheng People's Hospital, Weifang, Shandong 262200, P.R. China
| | - Yan Liu
- Department of Gastroenterology, Qiqihar Jianhua Hospital, Qiqihar, Heilongjiang 161000, P.R. China
| | - Weiguo Zhang
- Second Department of General Surgery, Tianjin Fifth Central Hospital, Tianjin 300450, P.R. China
| | - Tingting Ji
- Department of Gastroenterology, No. 215 Hospital of Shaanxi Nuclear Industry, Xianyang, Shaanxi 712000, P.R. China
| |
Collapse
|
15
|
Souza MDA, Ramos-Sanchez EM, Muxel SM, Lagos D, Reis LC, Pereira VRA, Brito MEF, Zampieri RA, Kaye PM, Floeter-Winter LM, Goto H. miR-548d-3p Alters Parasite Growth and Inflammation in Leishmania (Viannia) braziliensis Infection. Front Cell Infect Microbiol 2021; 11:687647. [PMID: 34178725 PMCID: PMC8224172 DOI: 10.3389/fcimb.2021.687647] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/24/2021] [Indexed: 12/21/2022] Open
Abstract
American Tegumentary Leishmaniasis (ATL) is an endemic disease in Latin America, mainly caused in Brazil by Leishmania (Viannia) braziliensis. Clinical manifestations vary from mild, localized cutaneous leishmaniasis (CL) to aggressive mucosal disease. The host immune response strongly determines the outcome of infection and pattern of disease. However, the pathogenesis of ATL is not well understood, and host microRNAs (miRNAs) may have a role in this context. In the present study, miRNAs were quantified using qPCR arrays in human monocytic THP-1 cells infected in vitro with L. (V.) braziliensis promastigotes and in plasma from patients with ATL, focusing on inflammatory response-specific miRNAs. Patients with active or self-healed cutaneous leishmaniasis patients, with confirmed parasitological or immunological diagnosis, were compared with healthy controls. Computational target prediction of significantly-altered miRNAs from in vitro L. (V.) braziliensis-infected THP-1 cells revealed predicted targets involved in diverse pathways, including chemokine signaling, inflammatory, cellular proliferation, and tissue repair processes. In plasma, we observed distinct miRNA expression in patients with self-healed and active lesions compared with healthy controls. Some miRNAs dysregulated during THP-1 in vitro infection were also found in plasma from self-healed patients, including miR-548d-3p, which was upregulated in infected THP-1 cells and in plasma from self-healed patients. As miR-548d-3p was predicted to target the chemokine pathway and inflammation is a central to the pathogenesis of ATL, we evaluated the effect of transient transfection of a miR-548d-3p inhibitor on L. (V.) braziliensis infected-THP-1 cells. Inhibition of miR-548d-3p reduced parasite growth early after infection and increased production of MCP1/CCL2, RANTES/CCL5, and IP10/CXCL10. In plasma of self-healed patients, MCP1/CCL2, RANTES/CCL5, and IL-8/CXCL8 concentrations were significantly decreased and MIG/CXCL9 and IP-10/CXCL10 increased compared to patients with active disease. These data suggest that by modulating miRNAs, L. (V.) braziliensis may interfere with chemokine production and hence the inflammatory processes underpinning lesion resolution. Our data suggest miR-548d-3p could be further evaluated as a prognostic marker for ATL and/or as a host-directed therapeutic target.
Collapse
Affiliation(s)
- Marina de Assis Souza
- Instituto de Medicina Tropical, Faculdade de Medicina, Universidade de São Paulo (IMTSP/USP), São Paulo, Brazil
| | - Eduardo Milton Ramos-Sanchez
- Instituto de Medicina Tropical, Faculdade de Medicina, Universidade de São Paulo (IMTSP/USP), São Paulo, Brazil.,Departamento de Salud Publica, Facultad de Ciencias de La Salud, Universidad Nacional Toribio Rodriguez de Mendoza de Amazonas, Chachapoyas, Peru
| | | | - Dimitris Lagos
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
| | - Luiza Campos Reis
- Instituto de Medicina Tropical, Faculdade de Medicina, Universidade de São Paulo (IMTSP/USP), São Paulo, Brazil
| | | | | | | | - Paul Martin Kaye
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
| | | | - Hiro Goto
- Instituto de Medicina Tropical, Faculdade de Medicina, Universidade de São Paulo (IMTSP/USP), São Paulo, Brazil.,Departamento de Medicina Preventiva, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
16
|
Paul S, Ruiz-Manriquez LM, Serrano-Cano FI, Estrada-Meza C, Solorio-Diaz KA, Srivastava A. Human microRNAs in host-parasite interaction: a review. 3 Biotech 2020; 10:510. [PMID: 33178551 PMCID: PMC7644590 DOI: 10.1007/s13205-020-02498-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 10/19/2020] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs (miRNAs) are a group of small noncoding RNA molecules with significant capacity to regulate the gene expression at the post-transcriptional level in a sequence-specific manner either through translation repression or mRNA degradation triggering a fine-tuning biological impact. They have been implicated in several processes, including cell growth and development, signal transduction, cell proliferation and differentiation, metabolism, apoptosis, inflammation, and immune response modulation. However, over the last few years, extensive studies have shown the relevance of miRNAs in human pathophysiology. Common human parasitic diseases, such as Malaria, Leishmaniasis, Amoebiasis, Chagas disease, Schistosomiasis, Toxoplasmosis, Cryptosporidiosis, Clonorchiasis, and Echinococcosis are the leading cause of death worldwide. Thus, identifying and characterizing parasite-specific miRNAs and their host targets, as well as host-related miRNAs, are important for a deeper understanding of the pathophysiology of parasite-specific diseases at the molecular level. In this review, we have demonstrated the impact of human microRNAs during host-parasite interaction as well as their potential to be used for diagnosis and prognosis purposes.
Collapse
Affiliation(s)
- Sujay Paul
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, 76130 Querétaro, Mexico
| | - Luis M. Ruiz-Manriquez
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, 76130 Querétaro, Mexico
| | - Francisco I. Serrano-Cano
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, 76130 Querétaro, Mexico
| | - Carolina Estrada-Meza
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, 76130 Querétaro, Mexico
| | - Karla A. Solorio-Diaz
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, 76130 Querétaro, Mexico
| | - Aashish Srivastava
- Section of Bioinformatics, Clinical Laboratory, Haukeland University Hospital, 5021 Bergen, Norway
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
| |
Collapse
|
17
|
Masoudzadeh N, Mizbani A, Rafati S. Transcriptomic profiling in Cutaneous Leishmaniasis patients. Expert Rev Proteomics 2020; 17:533-541. [PMID: 32886890 DOI: 10.1080/14789450.2020.1812390] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Cutaneous leishmaniasis (CL), caused by different Leishmania parasite species, is associated with parasite-induced immune-mediated skin inflammation and ulceration. Whereas many CL studies focus on gene expression signatures in mouse models, the transcriptional response driving human patients in the field is less characterized. Human studies in CL disease provide the opportunity to directly investigate the host-pathogen interaction in the cutaneous lesion site. AREAS COVERED Advances in high-throughput sequencing technologies, particularly their application for evaluation of the global gene expression changes, have made transcriptomics as a powerful tool to understand the pathogen-host molecular interactions. EXPERT COMMENTARY In this review, we focus on the transcriptomics studies that have been performed so far on human blood or tissue-driven samples to investigate Leishmania parasites interplay with the CL patients. Further, we summarize microarray and RNA-seq studies associated with lesion biopsies of CL patients to discuss how current whole genome analysis along with systems biology approaches have developed novel CL biomarkers for further applications, not only for research, but also for accelerating vaccine development.
Collapse
Affiliation(s)
- Nasrin Masoudzadeh
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran , Tehran, Iran
| | - Amir Mizbani
- Department of Health Sciences and Technology, ETH Zurich , Switzerland
| | - Sima Rafati
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran , Tehran, Iran
| |
Collapse
|
18
|
Zalewski DP, Ruszel KP, Stępniewski A, Gałkowski D, Bogucki J, Komsta Ł, Kołodziej P, Chmiel P, Zubilewicz T, Feldo M, Kocki J, Bogucka-Kocka A. Dysregulation of microRNA Modulatory Network in Abdominal Aortic Aneurysm. J Clin Med 2020; 9:jcm9061974. [PMID: 32599769 PMCID: PMC7355415 DOI: 10.3390/jcm9061974] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/13/2020] [Accepted: 06/22/2020] [Indexed: 12/16/2022] Open
Abstract
Abdominal artery aneurysm (AAA) refers to abdominal aortic dilatation of 3 cm or greater. AAA is frequently underdiagnosed due to often asymptomatic character of the disease, leading to elevated mortality due to aneurysm rupture. MiRNA constitute a pool of small RNAs controlling gene expression and is involved in many pathologic conditions in human. Targeted panel detecting altered expression of miRNA and genes involved in AAA would improve early diagnosis of this disease. In the presented study, we selected and analyzed miRNA and gene expression signatures in AAA patients. Next, generation sequencing was applied to obtain miRNA and gene-wide expression profiles from peripheral blood mononuclear cells in individuals with AAA and healthy controls. Differential expression analysis was performed using DESeq2 and uninformative variable elimination by partial least squares (UVE-PLS) methods. A total of 31 miRNAs and 51 genes were selected as the most promising biomarkers of AAA. Receiver operating characteristics (ROC) analysis showed good diagnostic ability of proposed biomarkers. Genes regulated by selected miRNAs were determined in silico and associated with functional terms closely related to cardiovascular and neurological diseases. Proposed biomarkers may be used for new diagnostic and therapeutic approaches in management of AAA. The findings will also contribute to the pool of knowledge about miRNA-dependent regulatory mechanisms involved in pathology of that disease.
Collapse
Affiliation(s)
- Daniel P. Zalewski
- Chair and Department of Biology and Genetics, Medical University of Lublin, 4a Chodźki St., 20-093 Lublin, Poland; (D.P.Z.); (P.C.)
| | - Karol P. Ruszel
- Chair of Medical Genetics, Department of Clinical Genetics, Medical University of Lublin, 11 Radziwiłłowska St., 20-080 Lublin, Poland; (K.P.R.); (J.B.); (J.K.)
| | - Andrzej Stępniewski
- Ecotech Complex Analytical and Programme Centre for Advanced Environmentally Friendly Technologies, University of Marie Curie-Skłodowska, 39 Głęboka St., 20-612 Lublin, Poland;
| | - Dariusz Gałkowski
- Department of Pathology and Laboratory Medicine, Rutgers - Robert Wood Johnson Medical School, One Robert Wood Johnson Place, New Brunswick, NJ 08903-0019, USA;
| | - Jacek Bogucki
- Chair of Medical Genetics, Department of Clinical Genetics, Medical University of Lublin, 11 Radziwiłłowska St., 20-080 Lublin, Poland; (K.P.R.); (J.B.); (J.K.)
| | - Łukasz Komsta
- Chair and Department of Medicinal Chemistry, Medical University of Lublin, 4 Jaczewskiego St., 20-090 Lublin, Poland;
| | - Przemysław Kołodziej
- Laboratory of Diagnostic Parasitology, Chair and Department of Biology and Genetics, Medical University of Lublin, 4a Chodźki St., 20-093 Lublin, Poland;
| | - Paulina Chmiel
- Chair and Department of Biology and Genetics, Medical University of Lublin, 4a Chodźki St., 20-093 Lublin, Poland; (D.P.Z.); (P.C.)
| | - Tomasz Zubilewicz
- Chair and Department of Vascular Surgery and Angiology, Medical University of Lublin, 11 Staszica St., 20-081 Lublin, Poland; (T.Z.); (M.F.)
| | - Marcin Feldo
- Chair and Department of Vascular Surgery and Angiology, Medical University of Lublin, 11 Staszica St., 20-081 Lublin, Poland; (T.Z.); (M.F.)
| | - Janusz Kocki
- Chair of Medical Genetics, Department of Clinical Genetics, Medical University of Lublin, 11 Radziwiłłowska St., 20-080 Lublin, Poland; (K.P.R.); (J.B.); (J.K.)
| | - Anna Bogucka-Kocka
- Chair and Department of Biology and Genetics, Medical University of Lublin, 4a Chodźki St., 20-093 Lublin, Poland; (D.P.Z.); (P.C.)
- Correspondence: ; Tel.: +48-81-448-7232
| |
Collapse
|
19
|
Huang K, Yu X, Yu Y, Zhang L, Cen Y, Chu J. Long noncoding RNA MALAT1 promotes high glucose-induced inflammation and apoptosis of vascular endothelial cells by regulating miR-361-3p/SOCS3 axis. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2020; 13:1243-1252. [PMID: 32509100 PMCID: PMC7270668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 04/14/2020] [Indexed: 06/11/2023]
Abstract
Vascular complications are the important pathophysiologic manifestations of patients with diabetes mellitus (DM) and many long non-coding RNAs (LncRNAs) are involved in this process. In this study, we aimed to investigate the relationships among LncRNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), microRNA-361-3p (miR-361-3p), and suppressor of cytokine signaling 3 (SOCS3) in high glucose (HG)-induced human umbilical vein endothelial cell (HUVEC) injury and its underlying mechanism. We found that HG treatment significantly promotes MALAT1 and SOCS3 expressions, but inhibits miR-361-3p expression in HUVECs. Furthermore, through bioinformatics analysis and dual luciferase assay, we found that MALAT1 directly sponges miR-361-3p to counteract its suppression on SOCS3 expression. Moreover, knockdown of MALAT1 evidently inhibits HG-induced inflammatory factors, including tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β) and IL-6 expressions in HUVECs (and HUVEC apoptosis) by regulating the miR-361-3p/SOCS3 axis. In conclusion, our results indicate that knockdown of MALAT1 inhibits HG-induced vascular endothelial injury through regulating miR-361-3p/SOCS3 axis, suggesting that inhibition of MALAT1 as a potential target for endothelial injury therapy for DM.
Collapse
Affiliation(s)
- Kai Huang
- Department of General Practice, Ningbo First Hospital Ningbo, P. R. China
| | - Xuxia Yu
- Department of General Practice, Ningbo First Hospital Ningbo, P. R. China
| | - Yushan Yu
- Department of General Practice, Ningbo First Hospital Ningbo, P. R. China
| | - Lu Zhang
- Department of General Practice, Ningbo First Hospital Ningbo, P. R. China
| | - Yin Cen
- Department of General Practice, Ningbo First Hospital Ningbo, P. R. China
| | - Jinguo Chu
- Department of General Practice, Ningbo First Hospital Ningbo, P. R. China
| |
Collapse
|
20
|
Hamrouni S, Bras-Gonçalves R, Kidar A, Aoun K, Chamakh-Ayari R, Petitdidier E, Messaoudi Y, Pagniez J, Lemesre JL, Meddeb-Garnaoui A. Design of multi-epitope peptides containing HLA class-I and class-II-restricted epitopes derived from immunogenic Leishmania proteins, and evaluation of CD4+ and CD8+ T cell responses induced in cured cutaneous leishmaniasis subjects. PLoS Negl Trop Dis 2020; 14:e0008093. [PMID: 32176691 PMCID: PMC7098648 DOI: 10.1371/journal.pntd.0008093] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 03/26/2020] [Accepted: 01/27/2020] [Indexed: 02/07/2023] Open
Abstract
Human leishmaniasis is a public health problem worldwide for which the development of a vaccine remains a challenge. T cell-mediated immune responses are crucial for protection. Peptide vaccines based on the identification of immunodominant T cell epitopes able to induce T cell specific immune responses constitute a promising strategy. Here, we report the identification of human leukocyte antigen class-I (HLA-I) and -II (HLA-II)-restricted multi-epitope peptides from Leishmania proteins that we have previously described as vaccine candidates. Promastigote Surface Antigen (PSA), LmlRAB (L. major large RAB GTPase) and Histone (H2B) were screened, in silico, for T cell epitopes. 6 HLA-I and 5 HLA-II-restricted multi-epitope peptides, able to bind to the most frequent HLA molecules, were designed and used as pools to stimulate PBMCs from individuals with healed cutaneous leishmaniasis. IFN-γ, IL-10, TNF-α and granzyme B (GrB) production was evaluated by ELISA/CBA. The frequency of IFN-γ-producing T cells was quantified by ELISpot. T cells secreting cytokines and memory T cells were analyzed by flow cytometry. 16 of 25 peptide pools containing HLA-I, HLA-II or HLA-I and -II peptides were able to induce specific and significant IFN-γ levels. No IL-10 was detected. 6 peptide pools were selected among those inducing the highest IFN-γ levels for further characterization. 3/6 pools were able to induce a significant increase of the percentages of CD4+IFN-γ+, CD8+IFN-γ+ and CD4+GrB+ T cells. The same pools also induced a significant increase of the percentages of bifunctional IFN-γ+/TNF-α+CD4+ and/or central memory T cells. We identified highly promiscuous HLA-I and -II restricted epitope combinations from H2B, PSA and LmlRAB proteins that stimulate both CD4+ and CD8+ T cell responses in recovered individuals. These multi-epitope peptides could be used as potential components of a polytope vaccine for human leishmaniasis. The control of leishmaniasis, a neglected tropical disease of public health importance, caused by protozoan parasites of the genus Leishmania, mainly relies on chemotherapy, which is highly toxic. Currently, there is no vaccine against human leishmaniasis. Peptide-based vaccines consisting of T cell epitopes identified within proteins of interest by epitope predictive algorithms are a promising strategy for vaccine development. Here, we identified multi-epitope peptides composed of HLA-I and -II-restricted epitopes, using immunoinformatic tools, within Leishmania proteins previously described as potential vaccine candidates. We showed that multi-epitope peptides used as pools were able to activate IFN-γ producing CD4+ as well as CD8+ T cells, both required for parasite elimination. In addition, granzyme B-producing CD4+ T cells, bifunctional CD4+ IFN-γ+/TNF-α+ and/or TNF-α+/IL-2+ T cells as well as CD4+ and CD8+ central memory T cells, all involved in Leishmania infection control, were significantly increased in response to multi-epitope peptide stimulation. As far as we know, no study has described the detection of both CD4+ and CD8+ T cell populations in response to stimulation by both HLA-I and II-restricted peptides in humans. The immunogenic HLA-I and -II-restricted multi-epitope peptides identified in this study could constitute potential vaccine candidates against human leishmaniasis.
Collapse
Affiliation(s)
- Sarra Hamrouni
- Laboratoire de Parasitologie Médicale, Biotechnologie et Biomolécules, Institut Pasteur de Tunis, Tunis, Tunisie
- Faculté des Sciences de Bizerte, Université de Carthage, Tunis, Tunisie
- UMR INTERTRYP, Université de Montpellier, IRD, CIRAD, Montpellier, France
| | | | | | - Karim Aoun
- Laboratoire de Parasitologie Médicale, Biotechnologie et Biomolécules, Institut Pasteur de Tunis, Tunis, Tunisie
| | - Rym Chamakh-Ayari
- Laboratoire de Parasitologie Médicale, Biotechnologie et Biomolécules, Institut Pasteur de Tunis, Tunis, Tunisie
- Faculté des Sciences de Bizerte, Université de Carthage, Tunis, Tunisie
| | - Elodie Petitdidier
- UMR INTERTRYP, Université de Montpellier, IRD, CIRAD, Montpellier, France
| | - Yasmine Messaoudi
- Laboratoire de Parasitologie Médicale, Biotechnologie et Biomolécules, Institut Pasteur de Tunis, Tunis, Tunisie
- Faculté des Sciences de Bizerte, Université de Carthage, Tunis, Tunisie
- UMR INTERTRYP, Université de Montpellier, IRD, CIRAD, Montpellier, France
| | - Julie Pagniez
- UMR INTERTRYP, Université de Montpellier, IRD, CIRAD, Montpellier, France
| | - Jean-Loup Lemesre
- UMR INTERTRYP, Université de Montpellier, IRD, CIRAD, Montpellier, France
| | - Amel Meddeb-Garnaoui
- Laboratoire de Parasitologie Médicale, Biotechnologie et Biomolécules, Institut Pasteur de Tunis, Tunis, Tunisie
- * E-mail:
| |
Collapse
|
21
|
Acuña SM, Floeter-Winter LM, Muxel SM. MicroRNAs: Biological Regulators in Pathogen-Host Interactions. Cells 2020; 9:E113. [PMID: 31906500 PMCID: PMC7016591 DOI: 10.3390/cells9010113] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/19/2019] [Accepted: 12/19/2019] [Indexed: 12/12/2022] Open
Abstract
An inflammatory response is essential for combating invading pathogens. Several effector components, as well as immune cell populations, are involved in mounting an immune response, thereby destroying pathogenic organisms such as bacteria, fungi, viruses, and parasites. In the past decade, microRNAs (miRNAs), a group of noncoding small RNAs, have emerged as functionally significant regulatory molecules with the significant capability of fine-tuning biological processes. The important role of miRNAs in inflammation and immune responses is highlighted by studies in which the regulation of miRNAs in the host was shown to be related to infectious diseases and associated with the eradication or susceptibility of the infection. Here, we review the biological aspects of microRNAs, focusing on their roles as regulators of gene expression during pathogen-host interactions and their implications in the immune response against Leishmania, Trypanosoma, Toxoplasma, and Plasmodium infectious diseases.
Collapse
Affiliation(s)
| | | | - Sandra Marcia Muxel
- Department of Physiology, Universidade de São Paulo, 05508-090 São Paulo, Brazil; (S.M.A.); (L.M.F.-W.)
| |
Collapse
|
22
|
El-Shal AS, Matboli M, Abdelaziz AM, Morsy AA, Abdelbary EH. Role of a novel circulatory RNA-based biomarker panel expression in ovarian cancer. IUBMB Life 2019; 71:2031-2047. [PMID: 31520466 DOI: 10.1002/iub.2153] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 08/06/2019] [Indexed: 12/20/2022]
Abstract
Ovarian cancer (OC) is considered the sixth commonest cancer affecting women globally. We choose novel integrated specific ovarian cancer RNA biomarker panel; pellino E3 ubiquitin protein ligase family member 3 (PELI3) gene expressions along with its selected epigenetic regulators (microRNA (miR-361-3p) and long noncoding RNA (lncRNA RP5-837J1.2) by bioinformatic methods. Then, differential expressions of the selected panel in the sera of 50 OC patients, 42 cases with benign ovarian lesions, and among 45 controls were determined using real-time polymerase chain reaction quantitative (qRT-PCR). Furthermore, their expression was measured also in malignant ovarian tissues and adjacent nontumor tissues in 23 of 50 OC patients by quantitative qRT-PCR. The current study reported, for the first time, upregulation of serum lncRNA RP5-837J1.2 with concomitant downregulation of miR-361-3p and PELI3 mRNA in malignant group compared with benign and controls groups. There were associations of serum lncRNA RP5-837J1.2 with the affected ovary and worse International Federation of Gynecology and Obstetrics staging; associations of miR-361-3p with tumor size, grade, stage, and presence of metastasis; as well as associations among PELI3 mRNA expression and tumor size, grade, stage, and presence of metastasis among the OC group. In tumor tissues, miR-361-3p and PELI3 mRNA levels were at a higher level than that of nontumor tissues; however, tumor tissue showed lower level of lncRNA RP5-837J1.2 compared to normal tissue. There were positive correlations between serum and tissue level of RNA RP5-837J1.2, miR-361-3p, and PELI3 mRNA, but they did not reach statistical significance. Receiver operating characteristics curve analyses showed that lncRNA RP5-837J1.2, miR-361-3p, and PELI3 mRNA expression levels can discriminate among OC patient, cases with benign mass, and controls with an accuracy of 96, 76, and 83%, respectively; which increased if they are combined. This novel diagnostic RNA-based panel biomarker could be helpful for OC diagnosis.
Collapse
Affiliation(s)
- Amal S El-Shal
- Medical Biochemistry Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Marwa Matboli
- Medical Biochemistry and Molecular biology Department, Faculty of Medicine, Ain Shams University Research Institute, Cairo, Egypt
| | - Ahmed M Abdelaziz
- Obstetrics and Gynecology Department, Faculty of Medicine, Benha University, Benha, Egypt
| | - Ali A Morsy
- Obstetrics and Gynecology Department, Faculty of Medicine, Benha University, Benha, Egypt
| | - Eman H Abdelbary
- Pathology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
23
|
Afrin F, Khan I, Hemeg HA. Leishmania-Host Interactions-An Epigenetic Paradigm. Front Immunol 2019; 10:492. [PMID: 30967861 PMCID: PMC6438953 DOI: 10.3389/fimmu.2019.00492] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 02/25/2019] [Indexed: 12/17/2022] Open
Abstract
Leishmaniasis is one of the major neglected tropical diseases, for which no vaccines exist. Chemotherapy is hampered by limited efficacy coupled with development of resistance and other side effects. Leishmania parasites elude the host defensive mechanisms by modulating their surface proteins as well as dampening the host's immune responses. The parasites use the conventional RNA polymerases peculiarly under different environmental cues or pressures such as the host's milieu or the drugs. The mechanisms that restructure post-translational modifications are poorly understood but altered epigenetic histone modifications are believed to be instrumental in influencing the chromatin remodeling in the parasite. Interestingly, the parasite also modulates gene expression of the hosts, thereby hijacking or dampening the host immune response. Epigenetic factor such as DNA methylation of cytosine residues has been incriminated in silencing of macrophage-specific genes responsible for defense against these parasites. Although there is dearth of information regarding the epigenetic alterations-mediated pathogenesis in these parasites and the host, the unique epigenetic marks may represent targets for potential anti-leishmanial drug candidates. This review circumscribes the epigenetic changes during Leishmania infection, and the epigenetic modifications they enforce upon the host cells to ensure a safe haven. The non-coding micro RNAs as post-transcriptional regulators and correlates of wound healing and toll-like receptor signaling, as well as prognostic biomarkers of therapeutic failure and healing time are also explored. Finally, we highlight the recent advances on how the epigenetic perturbations may impact leishmaniasis vaccine development as biomarkers of safety and immunogenicity.
Collapse
Affiliation(s)
- Farhat Afrin
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Taibah University, Madina, Saudi Arabia
| | - Inbesat Khan
- Rajiv Gandhi Technical University, Bhopal, India
| | - Hassan A Hemeg
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Taibah University, Madina, Saudi Arabia
| |
Collapse
|