1
|
Zhang H, Yan R, Liu Y, Yu M, He Z, Xiao J, Li K, Liu G, Ning Q, Li Y. Progress in antileishmanial drugs: Mechanisms, challenges, and prospects. PLoS Negl Trop Dis 2025; 19:e0012735. [PMID: 39752369 PMCID: PMC11698350 DOI: 10.1371/journal.pntd.0012735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
Abstract
Leishmaniasis, a neglected tropical disease caused by Leishmania parasites, continues to pose global health challenges. Current treatments face issues like resistance, safety, efficacy, and cost. This review covers the discovery, mechanisms of action, clinical applications, and limitations of key antileishmanial agents: pentavalent antimonials, amphotericin B, miltefosine, paromomycin, and pentamidine. Despite toxicity and resistance (antimonials), hospitalization needs and side effects (amphotericin B), regional efficacy variability (miltefosine), inconsistent outcomes (paromomycin), and severe side effects (pentamidine), these drugs are vital. Novel strategies to overcome the deficiencies of current therapies are highlighted, including combination regimens, advanced drug delivery systems, and immunomodulatory approaches. Comprehensive and cooperative efforts are crucial to fully realize the potential of advancements in antileishmanial pharmacotherapy and to reduce the unacceptable worldwide burden imposed by this neglected disease.
Collapse
Affiliation(s)
- Haoran Zhang
- Department of Infectious Disease, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Ruixi Yan
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Yahui Liu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Mengtao Yu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Ziyi He
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Junfeng Xiao
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Kaijie Li
- Hubei Provincial Center for Disease Control and Prevention, Wuhan, China
| | - Gang Liu
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Qin Ning
- Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Li
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
2
|
Fernandes JCR, Zamboni DS. Mechanisms regulating host cell death during Leishmania infection. mBio 2024; 15:e0198023. [PMID: 39392429 PMCID: PMC11559009 DOI: 10.1128/mbio.01980-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024] Open
Abstract
Parasites from the Leishmania genus are the causative agents of leishmaniasis and primarily reside within macrophages during mammalian infection. Their ability to establish intracellular infection provides a secure niche for proliferation while evading detection. However, successful multiplication within mammalian cells requires the orchestration of multiple mechanisms that control host cell viability. In contrast, innate immune cells, such as macrophages, can undergo different forms of cell death in response to pathogenic intracellular microbes. Thus, modulation of these different forms of host cell death is crucial for Leishmaniasis development. The regulation of host cell apoptosis, a form of programmed cell death, is crucial for sustaining parasites within viable host cells. Accordingly, several studies have demonstrated evasion of apoptosis induced by dermotropic and viscerotropic Leishmania species. Conversely, the prevention of pyroptosis, an inflammatory form of cell death, ensures the establishment of infection by silencing the release of mediators that could trigger massive proinflammatory responses. This manuscript explores how Leishmania regulates various host cell death pathways and overviews seminal studies on regulating host cell apoptosis by different Leishmania species.
Collapse
Affiliation(s)
- Juliane C. R. Fernandes
- Department of Cell Biology, School of Medicine of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Dario S. Zamboni
- Department of Cell Biology, School of Medicine of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
3
|
Ribeiro JM, Teixeira EDM, Alves LL, Alves ÉAR, Pascoal-Xavier MA, Santi AMM, Oliveira E, Guimarães PPG, Teixeira-Carvalho A, Murta SMF, Peruhype-Magalhães V, Souza-Fagundes EM. Can letrozole be repurposed for the treatment of visceral leishmaniasis? Antimicrob Agents Chemother 2024; 68:e0075624. [PMID: 39387580 PMCID: PMC11540148 DOI: 10.1128/aac.00756-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 09/06/2024] [Indexed: 10/15/2024] Open
Abstract
Visceral leishmaniasis, caused by Leishmania infantum in New World countries, is the most serious and potentially fatal form of leishmaniasis, if left untreated. There are currently no effective prophylactic measures, and therapeutic options are limited. Therefore, we investigated whether the aromatase inhibitor letrozole (LET), which is already used to treat breast cancer, has an antileishmanial activity and/or immunomodulatory potential and therefore may be used to treat L. infantum infection. LET was active against L. infantum promastigote and amastigote life cycle stages in an in vitro infection model using human THP-1 cell-derived macrophages. In human peripheral blood leukocytes ex vivo, LET reduced the internalized forms of L. infantum by classical monocytes and activated neutrophils. Concomitantly, LET stimulated the production of IL-12/TNF-α and decreased the production of IL-10/TGF-β by peripheral blood phagocytes, while in T and B cells, it promoted the production of TNF-α/IFN-γ and decreased that of IL-10. In a murine infection model, LET significantly reduced the parasite load in the liver after just 5 days and in the spleen after 15 days. During in vivo treatment with LET, the production of TNF-α/IFN-γ also increased. In addition, the proportion of developing granulomas decreased and that of mature granulomas increased in the liver, while there was no significant change in organ architecture in the spleen. Based on these data, repositioning of LET may be promising for the treatment of visceral leishmaniasis in humans.
Collapse
Affiliation(s)
- Juliana Martins Ribeiro
- Departamento de
Fisiologia e Biofísica, Instituto de Ciências
Biológicas, Universidade Federal de Minas
Gerais, Belo Horizonte,
Minas Gerais, Brazil
- Grupo de
Genômica Funcional de Parasitos, Instituto René Rachou,
Fundação Oswaldo Cruz,
Belo Horizonte, Minas Gerais,
Brazil
| | - Eliane de Morais Teixeira
- Grupo Pesquisa
Clínica e Políticas Públicas em Doenças
Infecto-Parasitárias, Instituto René Rachou,
Fundação Oswaldo Cruz,
Belo Horizonte, Minas Gerais,
Brazil
| | - Líndicy Leidicy Alves
- Grupo Pesquisa
Clínica e Políticas Públicas em Doenças
Infecto-Parasitárias, Instituto René Rachou,
Fundação Oswaldo Cruz,
Belo Horizonte, Minas Gerais,
Brazil
| | - Érica Alessandra Rocha Alves
- Grupo Imunologia
Celular e Molecular, Instituto René Rachou,
Fundação Oswaldo Cruz,
Belo Horizonte, Minas Gerais,
Brazil
| | | | - Ana Maria Murta Santi
- Departamento de
Fisiologia e Biofísica, Instituto de Ciências
Biológicas, Universidade Federal de Minas
Gerais, Belo Horizonte,
Minas Gerais, Brazil
| | - Edward Oliveira
- Grupo de
Genômica Funcional de Parasitos, Instituto René Rachou,
Fundação Oswaldo Cruz,
Belo Horizonte, Minas Gerais,
Brazil
| | - Pedro Pires Goulart Guimarães
- Departamento de
Fisiologia e Biofísica, Instituto de Ciências
Biológicas, Universidade Federal de Minas
Gerais, Belo Horizonte,
Minas Gerais, Brazil
| | - Andrea Teixeira-Carvalho
- Grupo Integrado de
Pesquisa em Biomarcadores, Instituto René Rachou,
Fundação Oswaldo Cruz,
Belo Horizonte, Minas Gerais,
Brazil
| | - Silvane Maria Fonseca Murta
- Grupo de
Genômica Funcional de Parasitos, Instituto René Rachou,
Fundação Oswaldo Cruz,
Belo Horizonte, Minas Gerais,
Brazil
| | - Vanessa Peruhype-Magalhães
- Grupo Integrado de
Pesquisa em Biomarcadores, Instituto René Rachou,
Fundação Oswaldo Cruz,
Belo Horizonte, Minas Gerais,
Brazil
| | - Elaine Maria Souza-Fagundes
- Departamento de
Fisiologia e Biofísica, Instituto de Ciências
Biológicas, Universidade Federal de Minas
Gerais, Belo Horizonte,
Minas Gerais, Brazil
| |
Collapse
|
4
|
Ihedioha OC, Marcarian HQ, Sivakoses A, Beverley SM, McMahon-Pratt D, Bothwell ALM. Leishmania major surface components and DKK1 signalling via LRP6 promote migration and longevity of neutrophils in the infection site. Front Immunol 2024; 15:1473133. [PMID: 39502693 PMCID: PMC11534728 DOI: 10.3389/fimmu.2024.1473133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 09/30/2024] [Indexed: 11/08/2024] Open
Abstract
Background Host-related factors highly regulate the increased circulation of neutrophils during Leishmania infection. Platelet-derived Dickkopf-1 (DKK1) is established as a high-affinity ligand to LRP6. Recently, we demonstrated that DKK1 upregulates leukocyte-platelet aggregation, infiltration of neutrophils to the draining lymph node and Th2 differentiation during Leishmania infection, suggesting the potential involvement of the DKK1-LRP6 signalling pathway in neutrophil migration in infectious diseases. Results In this study, we further explored the potential role of DKK1-LRP6 signalling in the migration and longevity of activated neutrophils in the infection site using BALB/c mice with PMNs deficient in LRP6 (LRP6NKO) or BALB/c mice deficient in both PMN LRP6 and platelet DKK1 (LRP6NKO DKK1PKO). Relative to the infected wild-type BALB/c mice, reduced neutrophil activation at the infection site of LRP6NKO or LRP6NKO DKK1PKO mice was noted. The neutrophils obtained from either infected LRP6NKO or LRP6NKO DKK1PKO mice additionally showed a high level of apoptosis. Notably, the level of LRP6 expressing neutrophils was elevated in infected BALB/c mice. Relative to infected BALB/c mice, a significant reduction in parasite load was observed in both LRP6NKO and LRP6NKO DKK1PKO infected mice. Notably, DKK1 levels were comparable in the LRP6NKO and BALB/c mice in response to infection, indicating that PMN activation is the major pathway for DKK1 in promoting parasitemia. Parasite-specific components also play a crucial role in modulating neutrophil circulation in Leishmania disease. Thus, we further determine the contribution of Leishmania membrane components in the migration of neutrophils to the infection site using null mutants deficient in LPG synthesis (Δlpg1- ) or lacking all ether phospholipids (plasmalogens, LPG, and GIPLs) synthesis (Δads1- ). Relative to the WT controls, Δads1- parasite-infected mice showed a sustained decrease in neutrophils and neutrophil-platelet aggregates (for at least 14 days PI), while neutrophils returned to normal in Δlpg1- parasite-infected mice after day 3 PI. Conclusion Our results suggest that DKK1 signalling and Leishmania pathogen-associated molecular patterns appear to regulate the migration and sustenance of viable activated neutrophils in the infection site resulting in chronic type 2 cell-mediated inflammation.
Collapse
Affiliation(s)
- Olivia C. Ihedioha
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Haley Q. Marcarian
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Anutr Sivakoses
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Stephen M. Beverley
- Department of Molecular Microbiology, Washington University School of Medicine in St Louis, St. Louis, MO, United States
| | - Diane McMahon-Pratt
- Department of Epidemiology of Infectious Diseases, Yale School of Public Health, New Haven, CT, United States
| | - Alfred L. M. Bothwell
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
5
|
Solano-Gálvez SG, Gutiérrez-Kobeh L, Wilkins-Rodríguez AA, Vázquez-López R. Artemisinin: An Anti-Leishmania Drug that Targets the Leishmania Parasite and Activates Apoptosis of Infected Cells. Arch Med Res 2024; 55:103041. [PMID: 38996535 DOI: 10.1016/j.arcmed.2024.103041] [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: 01/15/2024] [Revised: 06/11/2024] [Accepted: 06/28/2024] [Indexed: 07/14/2024]
Abstract
Leishmaniasis is a relevant disease worldwide due to its presence in many countries and an estimated prevalence of 10 million people. The causative agent of this disease is the obligate intracellular parasite Leishmania which can infect different cell types. Part of its success depends on its ability to evade host defense mechanisms such as apoptosis. Apoptosis is a finely programmed process of cell death in which cells silently dismantle and actively participate in several processes such as immune response, differentiation, and cell growth. Leishmania has the ability to delay its initiation to persist in the cell. It has been well documented that different Leishmania species target different pathways that lead to apoptosis of cells such as macrophages, neutrophils, and dendritic cells. In many cases, the observed anti-apoptotic effect has been associated with a significant reduction in caspase-3 activity. Leishmania has also been shown to target several pathways involved in apoptosis such as MAPK, PI3K/Akt, and the antiapoptotic protein Bcl-xL. Understanding the strategies used by Leishmania to subvert the defense mechanisms of host cells, particularly apoptosis, is very relevant for the development of therapies and vaccines. In recent years, the drug artemisinin has been shown to be effective against several parasitic diseases. Its role against Leishmania may be promising. In this review, we provide important aspects of the disease, the strategies used by the parasite to suppress apoptosis, and the role of artemisinin in Leishmania infection.
Collapse
Affiliation(s)
- Sandra Georgina Solano-Gálvez
- Unidad de Investigación, División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Laila Gutiérrez-Kobeh
- Unidad de Investigación, División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Arturo A Wilkins-Rodríguez
- Unidad de Investigación, División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Rosalino Vázquez-López
- Departamento de Microbiología, Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud Universidad Anáhuac México Norte, Huixquilucan, Estado de México, Mexico.
| |
Collapse
|
6
|
Palomino-Cano C, Moreno E, Irache JM, Espuelas S. Targeting and activation of macrophages in leishmaniasis. A focus on iron oxide nanoparticles. Front Immunol 2024; 15:1437430. [PMID: 39211053 PMCID: PMC11357945 DOI: 10.3389/fimmu.2024.1437430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/16/2024] [Indexed: 09/04/2024] Open
Abstract
Macrophages play a pivotal role as host cells for Leishmania parasites, displaying a notable functional adaptability ranging from the proinflammatory, leishmanicidal M1 phenotype to the anti-inflammatory, parasite-permissive M2 phenotype. While macrophages can potentially eradicate amastigotes through appropriate activation, Leishmania employs diverse strategies to thwart this activation and redirect macrophages toward an M2 phenotype, facilitating its survival and replication. Additionally, a competition for iron between the two entities exits, as iron is vital for both and is also implicated in macrophage defensive oxidative mechanisms and modulation of their phenotype. This review explores the intricate interplay between macrophages, Leishmania, and iron. We focus the attention on the potential of iron oxide nanoparticles (IONPs) as a sort of immunotherapy to treat some leishmaniasis forms by reprogramming Leishmania-permissive M2 macrophages into antimicrobial M1 macrophages. Through the specific targeting of iron in macrophages, the use of IONPs emerges as a promising strategy to finely tune the parasite-host interaction, endowing macrophages with an augmented antimicrobial arsenal capable of efficiently eliminating these intrusive microbes.
Collapse
Affiliation(s)
- Carmen Palomino-Cano
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
| | - Esther Moreno
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
| | - Juan M. Irache
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Navarra Medical Research Institute (IdiSNA), Pamplona, Spain
| | - Socorro Espuelas
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Navarra Medical Research Institute (IdiSNA), Pamplona, Spain
| |
Collapse
|
7
|
Bamra T, Shafi T, Das S, Kumar M, Das P. Leishmania donovani mevalonate kinase regulates host actin for inducing phagocytosis. Biochimie 2024; 220:31-38. [PMID: 38123120 DOI: 10.1016/j.biochi.2023.12.003] [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: 08/07/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023]
Abstract
Despite the well-established role of macrophages in phagocytosing Leishmania, the contribution of the parasite to this process is not well understood. Present study provides insights into the mechanism underlying the MVK-induced entry of L. donovani and improve our knowledge of host-pathogen interactions. We have discussed Mevalonate kinase (MVK)-induced actin reorganization, modulation of signaling pathways and host cell functions. Our results show that LdMVK gains access to macrophage cytosol and induces actin assembly modulation through the activation of actin-related proteins: VASP, Src and ERM. We have also demonstrated that LdMVK induces Ca2+ signaling and Akt pathway in macrophages, which are critical components of Leishmania survival and proliferation. Interestingly, we found that antibodies against LdMVK can kill Leishmania-infected macrophages in culture by forming extracellular traps, highlighting the potential of LdMVK in inhibiting parasite death. Overall, LdMVK is a virulent factor in Leishmania that mediates parasite internalization and host modulation by targeting host proteins phosphorylation and calcium homeostasis having significant implications in disease progression.
Collapse
Affiliation(s)
- Tanvir Bamra
- Department of Molecular Biology, ICMR- Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, Bihar, 800 007, India.
| | - Taj Shafi
- Department of Molecular Biology, ICMR- Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, Bihar, 800 007, India.
| | - Sushmita Das
- Department of Microbiology, All India Institute of Medical Sciences, Phulwarisharif, Patna, Bihar, 801 507, India.
| | - Manjay Kumar
- Department of Molecular Biology, ICMR- Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, Bihar, 800 007, India.
| | - Pradeep Das
- Department of Molecular Biology, ICMR- Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, Bihar, 800 007, India; Division of Parasitology, ICMR-National Institute of Cholera and Enteric Diseases, Beleghata, Kolkata, West Bengal, 700 010, India.
| |
Collapse
|
8
|
Reyaz E, Puri N, Selvapandiyan A. Global Remodeling of Host Proteome in Response to Leishmania Infection. ACS Infect Dis 2024; 10:5-19. [PMID: 38084821 DOI: 10.1021/acsinfecdis.3c00492] [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] [Indexed: 01/13/2024]
Abstract
The protozoan parasite Leishmania possesses an intrinsic ability to modulate a multitude of pathways in the host, toward aiding its own proliferation. In response, the host reprograms its cellular, immunological, and metabolic machinery to evade the parasite's lethal impact. Besides inducing various antioxidant signaling pathways to counter the elevated stress response proteins like heme oxygenase-1 (HO-1), Leishmania also attempts to delay host cell apoptosis by promoting anti-apoptotic proteins like Bcl-2. The downstream modulation of apoptotic proteins is regulated by effector pathways, including the PI3K/Akt survival pathway, the mitogen-activated protein kinases (MAPKs) signaling pathway, and STAT phosphorylation. In addition, Leishmania assists in its infection in a time-dependent manner by modulating the level of various proteins of autophagic machinery. Immune effector cells, such as mast cells and neutrophils, entrap and kill the pathogen by secreting various granular proteins. In contrast, the host macrophages exert their leishmanicidal effect by secreting various cytokines, such as IL-2, IL-12, etc. An interplay of various signaling pathways occurs in an organized network that is highly specific to both pathogen and host species. This Review analyzes the modulation of expression of proteins, including the cytokines, providing a realistic approach toward understanding the pathophysiology of disease and predicting some prominent markers for disease intervention and vaccine support strategies.
Collapse
Affiliation(s)
- Enam Reyaz
- Department of Molecular Medicine, Jamia Hamdard, New Delhi 110062, India
| | - Niti Puri
- School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | | |
Collapse
|
9
|
Rodríguez-González J, Gutiérrez-Kobeh L. Apoptosis and its pathways as targets for intracellular pathogens to persist in cells. Parasitol Res 2023; 123:60. [PMID: 38112844 PMCID: PMC10730641 DOI: 10.1007/s00436-023-08031-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 11/10/2023] [Indexed: 12/21/2023]
Abstract
Apoptosis is a finely programmed process of cell death in which cells silently dismantle and actively participate in several operations such as immune response, differentiation, and cell growth. It can be initiated by three main pathways: the extrinsic, the perforin granzyme, and the intrinsic that culminate in the activation of several proteins in charge of tearing down the cell. On the other hand, apoptosis represents an ordeal for pathogens that live inside cells and maintain a strong dependency with them; thus, they have evolved multiple strategies to manipulate host cell apoptosis on their behalf. It has been widely documented that diverse intracellular bacteria, fungi, and parasites can interfere with most steps of the host cell apoptotic machinery to inhibit or induce apoptosis. Indeed, the inhibition of apoptosis is considered a virulence property shared by many intracellular pathogens to ensure productive replication. Some pathogens intervene at an early stage by interfering with the sensing of extracellular signals or transduction pathways. Others sense cellular stress or target the apoptosis regulator proteins of the Bcl-2 family or caspases. In many cases, the exact molecular mechanisms leading to the interference with the host cell apoptotic cascade are still unknown. However, intense research has been conducted to elucidate the strategies employed by intracellular pathogens to modulate host cell death. In this review, we summarize the main routes of activation of apoptosis and present several processes used by different bacteria, fungi, and parasites to modulate the apoptosis of their host cells.
Collapse
Affiliation(s)
- Jorge Rodríguez-González
- Unidad de Investigación UNAM-INC, División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México-Instituto Nacional de Cardiología "Ignacio Chávez,", Juan Badiano No. 1, Col. Belisario Domínguez, Sección XVI, Delegación Tlalpan, C.P. 14080, Ciudad de México, México
- Laboratorio de Estudios Epidemiológicos, Clínicos, Diseños Experimentales e Investigación, Facultad de Ciencias Químicas, Universidad Autónoma "Benito Juárez" de Oaxaca, Oaxaca, Mexico
| | - Laila Gutiérrez-Kobeh
- Unidad de Investigación UNAM-INC, División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México-Instituto Nacional de Cardiología "Ignacio Chávez,", Juan Badiano No. 1, Col. Belisario Domínguez, Sección XVI, Delegación Tlalpan, C.P. 14080, Ciudad de México, México.
| |
Collapse
|
10
|
De-Leon-Lopez YS, Thompson ME, Kean JJ, Flaherty RA. The PI3K-Akt pathway is a multifaceted regulator of the macrophage response to diverse group B Streptococcus isolates. Front Cell Infect Microbiol 2023; 13:1258275. [PMID: 37928185 PMCID: PMC10622663 DOI: 10.3389/fcimb.2023.1258275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 09/25/2023] [Indexed: 11/07/2023] Open
Abstract
Group B Streptococcus (GBS), also known as Streptococcus agalactiae, is a common member of the microbial flora in healthy individuals. However, problems may arise when GBS-colonized mothers become pregnant. GBS may be transferred from a colonized mother to her newborn or developing fetus, which may result in complications such as miscarriage, pre-term birth, meningitis, pneumonia, or sepsis. Macrophages play an especially important role in the fetal and newborn response to GBS due to the limited development of the adaptive immune system early in life. The goal of this study was to expand what is currently known about how GBS manipulates macrophage cell signaling to evade the immune system and cause disease. To this end, we investigated whether the PI3K-Akt pathway was involved in several key aspects of the macrophage response to GBS. We explored whether certain GBS strains, such as sequence type (ST)-17 strains, rely on this pathway for the more rapid macrophage uptake they induce compared to other GBS strains. Our findings suggest that this pathway is, indeed, important for macrophage uptake of GBS. Consistent with these findings, we used immunofluorescence microscopy to demonstrate that more virulent strains of GBS induce more actin projections in macrophages than less virulent strains. Additionally, we explored whether PI3K-Akt signaling impacted the ability of GBS to survive within macrophages after phagocytosis and whether this pathway influenced the survival rate of macrophages themselves following GBS infection. The PI3K-Akt pathway was found to promote the survival of both macrophages and intracellular GBS following infection. We also observed that inhibition of the PI3K-Akt pathway significantly reduced GBS-mediated activation of NFκB, which is a key regulator of cell survival and inflammatory responses. Overall, these insights into strain-dependent GBS-mediated manipulation of the PI3K-Akt pathway and its downstream targets in infected macrophages may provide new insights for the development of diagnostic and therapeutic tools to combat severe GBS disease.
Collapse
Affiliation(s)
| | | | | | - Rebecca A. Flaherty
- Department of Biology and Health Science, Aquinas College, Grand Rapids, MI, United States
| |
Collapse
|
11
|
Pessôa-Pereira D, Scorza BM, Cyndari KI, Beasley EA, Petersen CA. Modulation of Macrophage Redox and Apoptotic Processes to Leishmania infantum during Coinfection with the Tick-Borne Bacteria Borrelia burgdorferi. Pathogens 2023; 12:1128. [PMID: 37764937 PMCID: PMC10537792 DOI: 10.3390/pathogens12091128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/01/2023] [Accepted: 09/02/2023] [Indexed: 09/29/2023] Open
Abstract
Canine leishmaniosis (CanL) is a zoonotic disease caused by protozoan Leishmania infantum. Dogs with CanL are often coinfected with tick-borne bacterial pathogens, including Borrelia burgdorferi in the United States. These coinfections have been causally associated with hastened disease progression and mortality. However, the specific cellular mechanisms of how coinfections affect microbicidal responses against L. infantum are unknown. We hypothesized that B. burgdorferi coinfection impacts host macrophage effector functions, prompting L. infantum intracellular survival. In vitro experiments demonstrated that exposure to B. burgdorferi spirochetes significantly increased L. infantum parasite burden and pro-inflammatory responses in DH82 canine macrophage cells. Induction of cell death and generation of mitochondrial ROS were significantly decreased in coinfected DH82 cells compared to uninfected and L. infantum-infected cells. Ex vivo stimulation of PBMCs from L. infantum-seronegative and -seropositive subclinical dogs with spirochetes and/or total Leishmania antigens promoted limited induction of IFNγ. Coexposure significantly induced expression of pro-inflammatory cytokines and chemokines associated with Th17 differentiation and neutrophilic and monocytic recruitment in PBMCs from L. infantum-seropositive dogs. Excessive pro-inflammatory responses have previously been shown to cause CanL pathology. This work supports effective tick prevention and risk management of coinfections as critical strategies to prevent and control L. infantum progression in dogs.
Collapse
Affiliation(s)
- Danielle Pessôa-Pereira
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA 52242, USA; (D.P.-P.); (B.M.S.); (E.A.B.)
- Center for Emerging Infectious Diseases, University of Iowa, Iowa City, IA 52242, USA;
| | - Breanna M. Scorza
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA 52242, USA; (D.P.-P.); (B.M.S.); (E.A.B.)
- Center for Emerging Infectious Diseases, University of Iowa, Iowa City, IA 52242, USA;
| | - Karen I. Cyndari
- Center for Emerging Infectious Diseases, University of Iowa, Iowa City, IA 52242, USA;
- Department of Emergency Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | - Erin A. Beasley
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA 52242, USA; (D.P.-P.); (B.M.S.); (E.A.B.)
- Center for Emerging Infectious Diseases, University of Iowa, Iowa City, IA 52242, USA;
| | - Christine A. Petersen
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA 52242, USA; (D.P.-P.); (B.M.S.); (E.A.B.)
- Center for Emerging Infectious Diseases, University of Iowa, Iowa City, IA 52242, USA;
| |
Collapse
|
12
|
Baars I, Jaedtka M, Dewitz LA, Fu Y, Franz T, Mohr J, Gintschel P, Berlin H, Degen A, Freier S, Rygol S, Schraven B, Kahlfuß S, van Zandbergen G, Müller AJ. Leishmania major drives host phagocyte death and cell-to-cell transfer depending on intracellular pathogen proliferation rate. JCI Insight 2023; 8:e169020. [PMID: 37310793 PMCID: PMC10443809 DOI: 10.1172/jci.insight.169020] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 06/05/2023] [Indexed: 06/15/2023] Open
Abstract
The virulence of intracellular pathogens relies largely on the ability to survive and replicate within phagocytes but also on release and transfer into new host cells. Such cell-to-cell transfer could represent a target for counteracting microbial pathogenesis. However, our understanding of the underlying cellular and molecular processes remains woefully insufficient. Using intravital 2-photon microscopy of caspase-3 activation in the Leishmania major-infected (L. major-infected) live skin, we showed increased apoptosis in cells infected by the parasite. Also, transfer of the parasite to new host cells occurred directly without a detectable extracellular state and was associated with concomitant uptake of cellular material from the original host cell. These in vivo findings were fully recapitulated in infections of isolated human phagocytes. Furthermore, we observed that high pathogen proliferation increased cell death in infected cells, and long-term residency within an infected host cell was only possible for slowly proliferating parasites. Our results therefore suggest that L. major drives its own dissemination to new phagocytes by inducing host cell death in a proliferation-dependent manner.
Collapse
Affiliation(s)
- Iris Baars
- Experimental Immunodynamics, Institute of Molecular and Clinical Immunology, Medical Faculty, and
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty and Center for Health and Medical Prevention (CHaMP), Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Moritz Jaedtka
- Division of Immunology, Paul Ehrlich Institute, Langen, Germany
- Institute for Immunology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Leon-Alexander Dewitz
- Experimental Immunodynamics, Institute of Molecular and Clinical Immunology, Medical Faculty, and
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty and Center for Health and Medical Prevention (CHaMP), Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Yan Fu
- Experimental Immunodynamics, Institute of Molecular and Clinical Immunology, Medical Faculty, and
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty and Center for Health and Medical Prevention (CHaMP), Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Tobias Franz
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty and Center for Health and Medical Prevention (CHaMP), Otto von Guericke University Magdeburg, Magdeburg, Germany
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Juliane Mohr
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty and Center for Health and Medical Prevention (CHaMP), Otto von Guericke University Magdeburg, Magdeburg, Germany
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Patricia Gintschel
- Experimental Immunodynamics, Institute of Molecular and Clinical Immunology, Medical Faculty, and
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty and Center for Health and Medical Prevention (CHaMP), Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Hannes Berlin
- Experimental Immunodynamics, Institute of Molecular and Clinical Immunology, Medical Faculty, and
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty and Center for Health and Medical Prevention (CHaMP), Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Angelina Degen
- Experimental Immunodynamics, Institute of Molecular and Clinical Immunology, Medical Faculty, and
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty and Center for Health and Medical Prevention (CHaMP), Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Sandra Freier
- Experimental Immunodynamics, Institute of Molecular and Clinical Immunology, Medical Faculty, and
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty and Center for Health and Medical Prevention (CHaMP), Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Stefan Rygol
- Experimental Immunodynamics, Institute of Molecular and Clinical Immunology, Medical Faculty, and
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty and Center for Health and Medical Prevention (CHaMP), Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Burkhart Schraven
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty and Center for Health and Medical Prevention (CHaMP), Otto von Guericke University Magdeburg, Magdeburg, Germany
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Sascha Kahlfuß
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty and Center for Health and Medical Prevention (CHaMP), Otto von Guericke University Magdeburg, Magdeburg, Germany
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Ger van Zandbergen
- Division of Immunology, Paul Ehrlich Institute, Langen, Germany
- Institute for Immunology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Andreas J. Müller
- Experimental Immunodynamics, Institute of Molecular and Clinical Immunology, Medical Faculty, and
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty and Center for Health and Medical Prevention (CHaMP), Otto von Guericke University Magdeburg, Magdeburg, Germany
- Helmholtz Centre for Infection Research, Braunschweig, Germany
| |
Collapse
|
13
|
Venugopal G, Bird JT, Roys H, Bowlin A, Fry L, Byrum SD, Weinkopff T. BOTH THE INFECTION STATUS AND INFLAMMATORY MICROENVIRONMENT INDUCE TRANSCRIPTIONAL REMODELING IN MACROPHAGES IN MURINE LEISHMANIAL LESIONS. J Parasitol 2023; 109:200-210. [PMID: 37270767 DOI: 10.1645/22-94] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023] Open
Abstract
Cutaneous leishmaniasis is caused by infection with the protozoan parasite Leishmania, which resides intracellularly in dermal macrophages (Mø), producing lesions. The skin lesions are characterized by proinflammatory cytokines and growth factors as well as inflammatory hypoxia, creating a stressful microenvironment for Mø. Of importance, not all Mø in lesions harbor parasites. To distinguish the influence of the parasite from the inflammatory microenvironment after Leishmania major (LM) infection on the Mø, we performed single-cell RNA sequencing and compared Mø associated with LM transcripts (or 'infected' Mø) with Mø not associated with LM transcripts (or 'bystander' Mø) within the lesions. Our findings show coordinated lysosomal expression and regulation signaling with increased cathepsin and H+-ATPase transcripts are upregulated in infected compared with bystander Mø. Additionally, eukaryotic initiation factor 2 (EIF2) signaling is downregulated in infected compared with bystander Mø, which includes many small and large ribosomal subunit (Rps and Rpl) transcripts being decreased in Mø harboring parasites. Furthermore, we also find EIF2 signaling including EIF, Rps, and Rpl transcripts being downregulated in bystander Mø compared with Mø from naïve skin. These data suggest that both the parasite and the inflammatory host microenvironment affect the transcription of ribosomal machinery in lesional Mø, thereby potentially affecting the ability of these cells to perform translation, protein synthesis, and thus function. Altogether, these results suggest that both the parasite and host inflammatory microenvironment independently drive transcriptional remodeling in Mø during LM infection in vivo.
Collapse
Affiliation(s)
- Gopinath Venugopal
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
| | - Jordan T Bird
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
- Arkansas Children's Research Institute, Little Rock, Arkansas 72202
| | - Hayden Roys
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
| | - Anne Bowlin
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
| | - Lucy Fry
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
| | - Stephanie D Byrum
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
- Arkansas Children's Research Institute, Little Rock, Arkansas 72202
| | - Tiffany Weinkopff
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
| |
Collapse
|
14
|
de Sá KSG, Amaral LA, Rodrigues TS, Ishimoto AY, de Andrade WAC, de Almeida L, Freitas-Castro F, Batah SS, Oliveira SC, Pastorello MT, Fabro AT, Zamboni DS. Gasdermin-D activation promotes NLRP3 activation and host resistance to Leishmania infection. Nat Commun 2023; 14:1049. [PMID: 36828815 PMCID: PMC9958042 DOI: 10.1038/s41467-023-36626-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/09/2023] [Indexed: 02/26/2023] Open
Abstract
Intracellular parasites from the Leishmania genus cause Leishmaniasis, a disease affecting millions of people worldwide. NLRP3 inflammasome is key for disease outcome, but the molecular mechanisms upstream of the inflammasome activation are still unclear. Here, we demonstrate that despite the absence of pyroptosis, Gasdermin-D (GSDMD) is active at the early stages of Leishmania infection in macrophages, allowing transient cell permeabilization, potassium efflux, and NLRP3 inflammasome activation. Further, GSDMD is processed into a non-canonical 25 kDa fragment. Gsdmd-/- macrophages and mice exhibit less NLRP3 inflammasome activation and are highly susceptible to infection by several Leishmania species, confirming the role of GSDMD for inflammasome-mediated host resistance. Active NLRP3 inflammasome and GSDMD are present in skin biopsies of patients, demonstrating activation of this pathway in human leishmaniasis. Altogether, our findings reveal that Leishmania subverts the normal functions of GSDMD, an important molecule to promote inflammasome activation and immunity in Leishmaniasis.
Collapse
Affiliation(s)
- Keyla S G de Sá
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Luana A Amaral
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Tamara S Rodrigues
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Adriene Y Ishimoto
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Warrison A C de Andrade
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Leticia de Almeida
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Felipe Freitas-Castro
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Sabrina S Batah
- Departamento de Patologia e Medicina Legal, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Sergio C Oliveira
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Mônica T Pastorello
- Departamento de Patologia e Medicina Legal, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Alexandre T Fabro
- Departamento de Patologia e Medicina Legal, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
- Serviço de Patologia do Hospital das Clínicas da Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Dario S Zamboni
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14049-900, Brazil.
| |
Collapse
|
15
|
Gupta D, Singh PK, Yadav PK, Narender T, Patil UK, Jain SK, Chourasia MK. Emerging strategies and challenges of molecular therapeutics in antileishmanial drug development. Int Immunopharmacol 2023; 115:109649. [PMID: 36603357 DOI: 10.1016/j.intimp.2022.109649] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 12/16/2022] [Accepted: 12/24/2022] [Indexed: 01/05/2023]
Abstract
Molecular therapy refers to targeted therapies based on molecules which have been intelligently directed towards specific biomolecular structures and include small molecule drugs, monoclonal antibodies, proteins and peptides, DNA or RNA-based strategies, targeted chemotherapy and nanomedicines. Molecular therapy is emerging as the most effective strategy to combat the present challenges of life-threatening visceral leishmaniasis, where the successful human vaccine is currently unavailable. Moreover, current chemotherapy-based strategies are associated with the issues of ineffective targeting, unavoidable toxicities, invasive therapies, prolonged treatment, high treatment costs and the development of drug-resistant strains. Thus, the rational approach to antileishmanial drug development primarily demands critical exploration and exploitation of biochemical differences between host and parasite biology, immunocharacteristics of parasite homing, and host-parasite interactions at the molecular/cellular level. Following this, the novel technology-based designing and development of host and/or parasite-targeted therapeutics having leishmanicidal and immunomodulatory activity is utmost essential to improve treatment efficacy. Thus, the present review is focused on immunological and molecular checkpoint targets in host-pathogen interaction, and molecular therapeutic prospects for Leishmania intervention, and the challenges ahead.
Collapse
Affiliation(s)
- Deepak Gupta
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar 470003, M.P., India; Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Pankaj K Singh
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India; Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad 500037, Telangana, India
| | - Pavan K Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Tadigoppula Narender
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Umesh K Patil
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar 470003, M.P., India
| | - Sanjay K Jain
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar 470003, M.P., India
| | - Manish K Chourasia
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India.
| |
Collapse
|
16
|
Ashley IA, Kitchen SA, Gorman LM, Grossman AR, Oakley CA, Suggett DJ, Weis VM, Rosset SL, Davy SK. Genomic conservation and putative downstream functionality of the phosphatidylinositol signalling pathway in the cnidarian-dinoflagellate symbiosis. Front Microbiol 2023; 13:1094255. [PMID: 36777026 PMCID: PMC9909359 DOI: 10.3389/fmicb.2022.1094255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 12/28/2022] [Indexed: 01/28/2023] Open
Abstract
The mutualistic cnidarian-dinoflagellate symbiosis underpins the evolutionary success of stony corals and the persistence of coral reefs. However, a molecular understanding of the signalling events that lead to the successful establishment and maintenance of this symbiosis remains unresolved. For example, the phosphatidylinositol (PI) signalling pathway has been implicated during the establishment of multiple mutualistic and parasitic interactions across the kingdoms of life, yet its role within the cnidarian-dinoflagellate symbiosis remains unexplored. Here, we aimed to confirm the presence and assess the specific enzymatic composition of the PI signalling pathway across cnidaria and dinoflagellates by compiling 21 symbiotic anthozoan (corals and sea anemones) and 28 symbiotic dinoflagellate (Symbiodiniaceae) transcriptomic and genomic datasets and querying genes related to this pathway. Presence or absence of PI-kinase and PI-phosphatase orthologs were also compared between a broad sampling of taxonomically related symbiotic and non-symbiotic species. Across the symbiotic anthozoans analysed, there was a complete and highly conserved PI pathway, analogous to the pathway found in model eukaryotes. The Symbiodiniaceae pathway showed similarities to its sister taxon, the Apicomplexa, with the absence of PI 4-phosphatases. However, conversely to Apicomplexa, there was also an expansion of homologs present in the PI5-phosphatase and PI5-kinase groups, with unique Symbiodiniaceae proteins identified that are unknown from non-symbiotic unicellular organisms. Additionally, we aimed to unravel the putative functionalities of the PI signalling pathway in this symbiosis by analysing phosphoinositide (PIP)-binding proteins. Analysis of phosphoinositide (PIP)-binding proteins showed that, on average, 2.23 and 1.29% of the total assemblies of anthozoan and Symbiodiniaceae, respectively, have the potential to bind to PIPs. Enrichment of Gene Ontology (GO) terms associated with predicted PIP-binding proteins within each taxon revealed a broad range of functions, including compelling links to processes putatively involved in symbiosis regulation. This analysis establishes a baseline for current understanding of the PI pathway across anthozoans and Symbiodiniaceae, and thus a framework to target future research.
Collapse
Affiliation(s)
- Immy A. Ashley
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
| | - Sheila A. Kitchen
- Department of Marine Biology, Texas A&M University at Galveston, Galveston, TX, United States
| | - Lucy M. Gorman
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
| | - Arthur R. Grossman
- Department of Plant Biology, The Carnegie Institution, Stanford, CA, United States
| | - Clinton A. Oakley
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
| | - David J. Suggett
- Climate Change Cluster, Faculty of Science, University of Technology Sydney, Broadway, NSW, Australia
| | - Virginia M. Weis
- Department of Integrative Biology, Oregon State University, Corvallis, OR, United States
| | - Sabrina L. Rosset
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
| | - Simon K. Davy
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand,*Correspondence: Simon K. Davy,
| |
Collapse
|
17
|
Guhe V, Ingale P, Tambekar A, Singh S. Systems biology of autophagy in leishmanial infection and its diverse role in precision medicine. Front Mol Biosci 2023; 10:1113249. [PMID: 37152895 PMCID: PMC10160387 DOI: 10.3389/fmolb.2023.1113249] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 04/05/2023] [Indexed: 05/09/2023] Open
Abstract
Autophagy is a contentious issue in leishmaniasis and is emerging as a promising therapeutic regimen. Published research on the impact of autophagic regulation on Leishmania survival is inconclusive, despite numerous pieces of evidence that Leishmania spp. triggers autophagy in a variety of cell types. The mechanistic approach is poorly understood in the Leishmania parasite as autophagy is significant in both Leishmania and the host. Herein, this review discusses the autophagy proteins that are being investigated as potential therapeutic targets, the connection between autophagy and lipid metabolism, and microRNAs that regulate autophagy and lipid metabolism. It also highlights the use of systems biology to develop novel autophagy-dependent therapeutics for leishmaniasis by utilizing artificial intelligence (AI), machine learning (ML), mathematical modeling, network analysis, and other computational methods. Additionally, we have shown many databases for autophagy and metabolism in Leishmania parasites that suggest potential therapeutic targets for intricate signaling in the autophagy system. In a nutshell, the detailed understanding of the dynamics of autophagy in conjunction with lipids and miRNAs unfolds larger dimensions for future research.
Collapse
|
18
|
Oliveira IH, Kjeldsen F, Melo-Braga MN, Verano-Braga T, de Andrade HM. Assessing the effects of Leishmania (Leishmania) infantum and L. (L.) amazonensis infections in macrophages using a quantitative proteome approach. Exp Parasitol 2022; 243:108413. [DOI: 10.1016/j.exppara.2022.108413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 09/19/2022] [Accepted: 10/18/2022] [Indexed: 11/30/2022]
|
19
|
Rashidi S, Mansouri R, Ali-Hassanzadeh M, Ghani E, Karimazar M, Muro A, Nguewa P, Manzano-Román R. miRNAs in the regulation of mTOR signaling and host immune responses: The case of Leishmania infections. Acta Trop 2022; 231:106431. [PMID: 35367408 DOI: 10.1016/j.actatropica.2022.106431] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 03/23/2022] [Accepted: 03/26/2022] [Indexed: 11/01/2022]
Abstract
Micro RNAs (miRNAs), as regulators of gene expression at the post-transcriptional level, can respond to/or interact with cell signaling and affect the pathogenesis of different diseases/infections. The interaction/crosstalk of miRNAs with various cellular signaling networks including mTOR (as a master regulator of signaling relevant to different cellular mechanisms) might lead to the initiation, progression or restriction of certain disease processes. There are numerous studies that have identified the crosstalk between regulatory miRNA expression and the mTOR pathway (or mTOR signaling regulated by miRNAs) in different diseases which has a dual function in pathogenesis. However, the corresponding information in parasitic infections remains scarce. miRNAs have been suggested as specific targets for therapeutic strategies in several disorders such as parasitic infections. Thus, the targeting of miRNAs (as the modulators/regulators of mTOR) by small molecules and RNA-based therapeutics and consequently managing and modulating mTOR signaling and the downstream/related cell signaling/pathways might shed some light on the design of new therapeutic strategies against parasitic diseases, including Leishmaniasis. Accordingly, the present study attempts to highlight the importance of the crosstalk between regulatory miRNAs and mTOR signaling, and to review the relevant insights into parasitic infections by focusing specifically on Leishmania.
Collapse
|
20
|
Kumar R, Kushawaha PK. Interferon inducible guanylate binding protein 1 restricts the growth of Leishmania donovani by modulating the level of cytokines/chemokines and MAP kinase transcription factors. Microb Pathog 2022; 168:105568. [DOI: 10.1016/j.micpath.2022.105568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/01/2022] [Accepted: 05/04/2022] [Indexed: 11/27/2022]
|
21
|
Seth A, Kar S. Host-directed antileishmanial interventions: Harvesting unripe fruits to reach fruition. Int Rev Immunol 2022; 42:217-236. [PMID: 35275772 DOI: 10.1080/08830185.2022.2047670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Leishmaniasis is an exemplary paradigm of immune evasion, fraught with the perils of limited clinical assistance, escalating costs of treatment and made worse with the lack of suitable vaccine. While drugs remain central to large-scale disease control, the growing emergence of parasite resistance necessitates the need for combination therapy involving host-directed immunological agents. Also, since prolonged disease progression is associated with strong immune suppression of the host, augmentation of host immunity via restoration of the immunoregulatory circuit involving antigen-presenting cells and T-cells, activation of macrophage function and/or CD4+ T helper 1 cell differentiation may serve as an ideal approach to resolve severe cases of leishmaniasis. As such, therapies that embody a synergistic approach that involve direct killing of the parasite in addition to elevating host immunity are likely to pave the way for widespread elimination of leishmaniasis in the future. With this review, we aim to recapitulate the various immunotherapeutic agents found to hold promise in antileishmanial treatment both in vitro and in vivo. These include parasite-specific antigens, dendritic cell-targeted therapy, recombinant inhibitors of various components intrinsic to immune cell signaling and agonists or antagonists to immune cells and cytokines. We also summarize their abilities to direct therapeutic skewing of the host cell-immune response and review their potential to combat the disease either alone, or as adjunct modalities.
Collapse
Affiliation(s)
- Anuradha Seth
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Lucknow, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Susanta Kar
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Lucknow, India
| |
Collapse
|
22
|
Bichiou H, Rabhi S, Ben Hamda C, Bouabid C, Belghith M, Piquemal D, Trentin B, Rabhi I, Guizani-Tabbane L. Leishmania Parasites Differently Regulate Antioxidant Genes in Macrophages Derived From Resistant and Susceptible Mice. Front Cell Infect Microbiol 2021; 11:748738. [PMID: 34722338 PMCID: PMC8554229 DOI: 10.3389/fcimb.2021.748738] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 09/20/2021] [Indexed: 12/30/2022] Open
Abstract
Macrophage-Leishmania interactions are central to parasite growth and disease outcome. Macrophages have developed various strategies to fight invaders, including oxidative burst. While some microorganisms seem to survive and even thrive in an oxidative environment, others are susceptible and get killed. To counter oxidative stress, macrophages switch the expressions of cytoprotective and detoxifying enzymes, which are downstream targets of the nuclear factor erythroid 2-related factor 2 (Nrf2), to enhance cell survival. We have explored the transcription of NRF2 and of its target genes and compared the effect of the parasite on their transcription in bone marrow-derived macrophages (BMdMs) from Leishmania-resistant and Leishmania-susceptible mice. While heme oxygenase 1 (HO-1) transcription is independent of the genetic background, the transcription of glutathione reductase (Gsr) and of cysteine/glutamate exchange transporter (Slc7a11), involved in glutathione accumulation, was differentially regulated in BMdMs from both mouse strains. We also show that, except for HO-1, known to favor the survival of the parasite, the transcription of the selected genes, including Gsr, CD36, and catalase (CAT), was actively repressed, if not at all time points at least at the later ones, by the parasite, especially in Balb/c BMdMs. Consistent with these results, we found that the silencing of NRF2 in this study increases the survival and multiplication of the parasite.
Collapse
Affiliation(s)
- Haifa Bichiou
- Laboratory of Medical Parasitology, Biotechnology and Biomolecules, Institut Pasteur de Tunis, Tunis-Belvedere, Tunisia.,Faculty of Sciences of Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Sameh Rabhi
- Laboratory of Medical Parasitology, Biotechnology and Biomolecules, Institut Pasteur de Tunis, Tunis-Belvedere, Tunisia
| | - Cherif Ben Hamda
- Laboratory of Medical Parasitology, Biotechnology and Biomolecules, Institut Pasteur de Tunis, Tunis-Belvedere, Tunisia
| | - Cyrine Bouabid
- Laboratory of Medical Parasitology, Biotechnology and Biomolecules, Institut Pasteur de Tunis, Tunis-Belvedere, Tunisia.,Faculty of Sciences of Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Meriam Belghith
- Department of Immunology, Institut Pasteur de Tunis, University Tunis El-Manar, Tunis, Tunisia
| | | | | | - Imen Rabhi
- Laboratory of Medical Parasitology, Biotechnology and Biomolecules, Institut Pasteur de Tunis, Tunis-Belvedere, Tunisia.,Higher Institute of Biotechnology at Sidi-Thabet, Biotechpole Sidi-Thabet, University of Manouba, Sidi-Thabet, Tunisia
| | - Lamia Guizani-Tabbane
- Laboratory of Medical Parasitology, Biotechnology and Biomolecules, Institut Pasteur de Tunis, Tunis-Belvedere, Tunisia
| |
Collapse
|
23
|
Bosurgi L, Rothlin CV. Management of cell death in parasitic infections. Semin Immunopathol 2021; 43:481-492. [PMID: 34279684 PMCID: PMC8443503 DOI: 10.1007/s00281-021-00875-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 05/27/2021] [Indexed: 12/15/2022]
Abstract
For a long time, host cell death during parasitic infection has been considered a reflection of tissue damage, and often associated with disease pathogenesis. However, during their evolution, protozoan and helminth parasites have developed strategies to interfere with cell death so as to spread and survive in the infected host, thereby ascribing a more intriguing role to infection-associated cell death. In this review, we examine the mechanisms used by intracellular and extracellular parasites to respectively inhibit or trigger programmed cell death. We further dissect the role of the prototypical “eat-me signal” phosphatidylserine (PtdSer) which, by being exposed on the cell surface of damaged host cells as well as on some viable parasites via a process of apoptotic mimicry, leads to their recognition and up-take by the neighboring phagocytes. Although barely dissected so far, the engagement of different PtdSer receptors on macrophages, by shaping the host immune response, affects the overall infection outcome in models of both protozoan and helminth infections. In this scenario, further understanding of the molecular and cellular regulation of the PtdSer exposing cell-macrophage interaction might allow the identification of new therapeutic targets for the management of parasitic infection.
Collapse
Affiliation(s)
- Lidia Bosurgi
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20251, Hamburg, Germany. .,Protozoa Immunology, Bernhard Nocht Institute for Tropical Medicine, Bernhard Nocht Strasse 74, 20359, Hamburg, Germany.
| | - Carla V Rothlin
- Department of Immunobiology and Pharmacology, Yale University, New Haven, CT, USA
| |
Collapse
|
24
|
Poulaki A, Piperaki ET, Voulgarelis M. Effects of Visceralising Leishmania on the Spleen, Liver, and Bone Marrow: A Pathophysiological Perspective. Microorganisms 2021; 9:microorganisms9040759. [PMID: 33916346 PMCID: PMC8066032 DOI: 10.3390/microorganisms9040759] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 03/30/2021] [Accepted: 04/01/2021] [Indexed: 01/29/2023] Open
Abstract
The leishmaniases constitute a group of parasitic diseases caused by species of the protozoan genus Leishmania. In humans it can present different clinical manifestations and are usually classified as cutaneous, mucocutaneous, and visceral (VL). Although the full range of parasite—host interactions remains unclear, recent advances are improving our comprehension of VL pathophysiology. In this review we explore the differences in VL immunobiology between the liver and the spleen, leading to contrasting infection outcomes in the two organs, specifically clearance of the parasite in the liver and failure of the spleen to contain the infection. Based on parasite biology and the mammalian immune response, we describe how hypoxia-inducible factor 1 (HIF1) and the PI3K/Akt pathway function as major determinants of the observed immune failure. We also summarize existing knowledge on pancytopenia in VL, as a direct effect of the parasite on bone marrow health and regenerative capacity. Finally, we speculate on the possible effect that manipulation by the parasite of the PI3K/Akt/HIF1 axis may have on the myelodysplastic (MDS) features observed in VL.
Collapse
Affiliation(s)
- Aikaterini Poulaki
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, 115 27 Athens, Greece;
| | - Evangelia-Theophano Piperaki
- Department of Microbiology, School of Medicine, National and Kapodistrian University of Athens, 115 27 Athens, Greece
- Correspondence: (E.-T.P.); (M.V.); Tel.: +30-210-7462136 (E.-T.P.); +30-210-7462647 (M.V.)
| | - Michael Voulgarelis
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, 115 27 Athens, Greece;
- Correspondence: (E.-T.P.); (M.V.); Tel.: +30-210-7462136 (E.-T.P.); +30-210-7462647 (M.V.)
| |
Collapse
|
25
|
Chandrakar P, Seth A, Rani A, Dutta M, Parmar N, Descoteaux A, Kar S. Jagged-Notch-mediated divergence of immune cell crosstalk maintains the anti-inflammatory response in visceral leishmaniasis. J Cell Sci 2021; 134:jcs.252494. [PMID: 33589499 DOI: 10.1242/jcs.252494] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 01/25/2021] [Indexed: 01/28/2023] Open
Abstract
Notch signaling governs crucial aspects of intercellular communication spanning antigen-presenting cells and T-cells. In this study, we investigate how Leishmania donovani takes advantage of this pathway to quell host immune responses. We report induction of the Notch ligand Jagged1 in L. donovani-infected bone marrow macrophages (BMMϕs) and subsequent activation of RBPJκ (also known as RBPJ) in T cells, which in turn upregulates the transcription factor GATA3. Activated RBPJκ also associates with the histone acetyltransferase p300 (also known as EP300), which binds with the Bcl2l12 promoter and enhances its expression. Interaction of Bcl2L12 with GATA3 in CD4+ T cells facilitates its binding to the interleukin (IL)-10 and IL-4 promoters, thereby increasing the secretion of these cytokines. Silencing Jagged1 hindered these events in a BMMϕ-T cell co-culture system. Upon further scrutiny, we found that parasite lipophosphoglycan (LPG) induces the host phosphoinositide 3-kinase (PI3K)/Akt pathway, which activates β-catenin and Egr1, the two transcription factors responsible for driving Jagged1 expression. In v ivo morpholino-silencing of Jagged1 suppresses anti-inflammatory cytokine responses and reduces organ parasite burden in L. donovani-infected Balb/c mice, suggesting that L. donovani-induced host Jagged1-Notch signaling skews macrophage-T cell crosstalk into disease-promoting Th2 mode in experimental visceral leishmaniasis.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Pragya Chandrakar
- Division of Molecular Parasitology and Immunology, CSIR-Central Drug Research Institute, Lucknow 226031, India.,Division of Biological Sciences, Academy of Scientific and Innovative Research (AcSIR), CSIR Human Resource Development Centre (CSIR-HRDC) Campus, Postal Staff College Area, Sector 19, Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh 201 002, India
| | - Anuradha Seth
- Division of Molecular Parasitology and Immunology, CSIR-Central Drug Research Institute, Lucknow 226031, India.,Division of Biological Sciences, Academy of Scientific and Innovative Research (AcSIR), CSIR Human Resource Development Centre (CSIR-HRDC) Campus, Postal Staff College Area, Sector 19, Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh 201 002, India
| | - Ankita Rani
- Division of Molecular Parasitology and Immunology, CSIR-Central Drug Research Institute, Lucknow 226031, India.,Division of Biological Sciences, Academy of Scientific and Innovative Research (AcSIR), CSIR Human Resource Development Centre (CSIR-HRDC) Campus, Postal Staff College Area, Sector 19, Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh 201 002, India
| | - Mukul Dutta
- Division of Molecular Parasitology and Immunology, CSIR-Central Drug Research Institute, Lucknow 226031, India.,Division of Biological Sciences, Academy of Scientific and Innovative Research (AcSIR), CSIR Human Resource Development Centre (CSIR-HRDC) Campus, Postal Staff College Area, Sector 19, Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh 201 002, India
| | - Naveen Parmar
- Division of Molecular Parasitology and Immunology, CSIR-Central Drug Research Institute, Lucknow 226031, India.,Division of Biological Sciences, Academy of Scientific and Innovative Research (AcSIR), CSIR Human Resource Development Centre (CSIR-HRDC) Campus, Postal Staff College Area, Sector 19, Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh 201 002, India
| | - Albert Descoteaux
- Centre for Host-Parasite Interactions, Institut National de la Recherche Scientifique-Centre Armand-Frappier Santé Biotechnologie, Laval, Quebec H7V 1B7, Canada
| | - Susanta Kar
- Division of Molecular Parasitology and Immunology, CSIR-Central Drug Research Institute, Lucknow 226031, India .,Division of Biological Sciences, Academy of Scientific and Innovative Research (AcSIR), CSIR Human Resource Development Centre (CSIR-HRDC) Campus, Postal Staff College Area, Sector 19, Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh 201 002, India
| |
Collapse
|
26
|
Solano-Gálvez SG, Álvarez-Hernández DA, Gutiérrez-Kobeh L, Vázquez-López R. Leishmania: manipulation of signaling pathways to inhibit host cell apoptosis. Ther Adv Infect Dis 2021; 8:20499361211014977. [PMID: 34104433 PMCID: PMC8165860 DOI: 10.1177/20499361211014977] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 04/13/2021] [Indexed: 11/17/2022] Open
Abstract
The maintenance of homeostasis in living systems requires the elimination of unwanted cells which is performed, among other mechanisms, by type I cell death or apoptosis. This type of programmed cell death involves several morphological changes such as cytoplasm shrinkage, chromatin condensation (pyknosis), nuclear fragmentation (karyorrhexis), and plasma membrane blebbing that culminate with the formation of apoptotic bodies. In addition to the maintenance of homeostasis, apoptosis also represents an important defense mechanism for cells against intracellular microorganisms. In counterpart, diverse intracellular pathogens have developed a wide array of strategies to evade apoptosis and persist inside cells. These strategies include the manipulation of signaling pathways involved in the inhibition of apoptosis where mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K) play a key role. Leishmania is an intracellular protozoan parasite that causes a wide spectrum of diseases known as leishmaniasis. This parasite displays different strategies, including apoptosis inhibition, to down-regulate host cell defense mechanisms in order to perpetuate infection.
Collapse
Affiliation(s)
- Sandra-Georgina Solano-Gálvez
- Unidad de Investigación UNAM-INC, División Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México, Instituto Nacional de Cardiología, Mexico City, Mexico
| | - Diego-Abelardo Álvarez-Hernández
- Departamento de Microbiología, Centro de Investigación en Ciencias de la Salud, CICSA Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte, Huixquilucán Estado de México, México
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, Bloomsbury, London, UK
| | - Laila Gutiérrez-Kobeh
- Unidad de Investigación UNAM-INC, División Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México, Instituto Nacional de Cardiología, Mexico City, Mexico
| | - Rosalino Vázquez-López
- Departamento de Microbiología, Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte, Av. Universidad Anáhuac 46, Col. Lomas Anáhuac, Huixquilucán Estado de México, Naucalpan, 52786, México
| |
Collapse
|
27
|
Rosset SL, Oakley CA, Ferrier-Pagès C, Suggett DJ, Weis VM, Davy SK. The Molecular Language of the Cnidarian-Dinoflagellate Symbiosis. Trends Microbiol 2020; 29:320-333. [PMID: 33041180 DOI: 10.1016/j.tim.2020.08.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/21/2020] [Accepted: 08/27/2020] [Indexed: 12/18/2022]
Abstract
The cnidarian-dinoflagellate symbiosis is of huge importance as it underpins the success of coral reefs, yet we know very little about how the host cnidarian and its dinoflagellate endosymbionts communicate with each other to form a functionally integrated unit. Here, we review the current knowledge of interpartner molecular signaling in this symbiosis, with an emphasis on lipids, glycans, reactive species, biogenic volatiles, and noncoding RNA. We draw upon evidence of these compounds from recent omics-based studies of cnidarian-dinoflagellate symbiosis and discuss the signaling roles that they play in other, better-studied symbioses. We then consider how improved knowledge of interpartner signaling might be used to develop solutions to the coral reef crisis by, for example, engineering more thermally resistant corals.
Collapse
Affiliation(s)
- Sabrina L Rosset
- School of Biological Sciences, Victoria University of Wellington, Wellington 6140, New Zealand
| | - Clinton A Oakley
- School of Biological Sciences, Victoria University of Wellington, Wellington 6140, New Zealand
| | | | - David J Suggett
- University of Technology Sydney, Climate Change Cluster, Faculty of Science, PO Box 123, Broadway NSW 2007, Australia
| | - Virginia M Weis
- Department of Integrative Biology, Oregon State University, Corvallis, OR 97331, USA
| | - Simon K Davy
- School of Biological Sciences, Victoria University of Wellington, Wellington 6140, New Zealand.
| |
Collapse
|
28
|
Lohrmann F, Forde AJ, Merck P, Henneke P. Control of myeloid cell density in barrier tissues. FEBS J 2020; 288:405-426. [PMID: 32502309 DOI: 10.1111/febs.15436] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 04/21/2020] [Accepted: 06/01/2020] [Indexed: 12/19/2022]
Abstract
The interface between the mammalian host and its environment is formed by barrier tissues, for example, of the skin, and the respiratory and the intestinal tracts. On the one hand, barrier tissues are colonized by site-adapted microbial communities, and on the other hand, they contain specific myeloid cell networks comprising macrophages, dendritic cells, and granulocytes. These immune cells are tightly regulated in function and cell number, indicating important roles in maintaining tissue homeostasis and immune balance in the presence of commensal microorganisms. The regulation of myeloid cell density and activation involves cell-autonomous 'single-loop circuits' including autocrine mechanisms. However, an array of microenvironmental factors originating from nonimmune cells and the microbiota, as well as the microanatomical structure, impose additional layers of regulation onto resident myeloid cells. This review discusses models integrating these factors into cell-specific programs to instruct differentiation and proliferation best suited for the maintenance and renewal of immune homeostasis in the tissue-specific environment.
Collapse
Affiliation(s)
- Florens Lohrmann
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine, Medical Center - University of Freiburg, Germany.,Institute for Immunodeficiency (IFI), Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, Germany.,Spemann Graduate School for Biology and Medicine, University of Freiburg, Germany.,IMM-PACT Clinician Scientist Program, Faculty of Medicine, University of Freiburg, Germany
| | - Aaron J Forde
- Institute for Immunodeficiency (IFI), Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, Germany.,Faculty of Biology, university of Freiburg, Germany
| | - Philipp Merck
- Institute for Immunodeficiency (IFI), Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, Germany
| | - Philipp Henneke
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine, Medical Center - University of Freiburg, Germany.,Institute for Immunodeficiency (IFI), Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, Germany
| |
Collapse
|
29
|
Crentsil JA, Yamthe LRT, Anibea BZ, Broni E, Kwofie SK, Tetteh JKA, Osei-Safo D. Leishmanicidal Potential of Hardwickiic Acid Isolated From Croton sylvaticus. Front Pharmacol 2020; 11:753. [PMID: 32523532 PMCID: PMC7261830 DOI: 10.3389/fphar.2020.00753] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 05/06/2020] [Indexed: 01/31/2023] Open
Abstract
Leishmania is a parasitic protozoon responsible for the neglected tropical disease Leishmaniasis. Approximately, 350 million people are susceptible and close to 70,000 death cases globally are reported annually. The lack of effective leishmanicides, the emergence of drug resistance and toxicity concerns necessitate the pursuit for effective antileishmanial drugs. Natural compounds serve as reservoirs for discovering new drugs due to their chemical diversity. Hardwickiic acid (HA) isolated from the stembark of Croton sylvaticus was evaluated for its leishmanicidal potential against Leishmania donovani and L. major promastigotes. The susceptibility of the promastigotes to HA was determined using the 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide/phenazine methosulfate colorimetric assay with Amphotericin B serving as positive control. HA showed a significant antileishmanial activity on L. donovani promastigotes with an IC50 value of 31.57± 0.06 µM with respect to the control drug, amphotericin B with IC50 of 3.35 ± 0.14 µM). The cytotoxic activity was observed to be CC50 = 247.83 ± 6.32 µM against 29.99 ± 2.82 µM for curcumin, the control, resulting in a selectivity index of SI = 7.85. Molecular modeling, docking and dynamics simulations of selected drug targets corroborated the observed antileishmanial activity of HA. Novel insights into the mechanisms of binding were obtained for trypanothione reductase (TR), pteridine reductase 1 (PTR1), and glutamate cysteine ligase (GCL). The binding affinity of HA to the drug targets LmGCL, LmPTR1, LdTR, LmTR, LdGCL, and LdPTR1 were obtained as -8.0, -7.8, -7.6, -7.5, -7.4 and -7.1 kcal/mol, respectively. The role of Lys16, Ser111, and Arg17 as critical residues required for binding to LdPTR1 was reinforced. HA was predicted as a Caspase-3 stimulant and Caspase-8 stimulant, implying a possible role in apoptosis, which was shown experimentally that HA induced parasite death by loss of membrane integrity. HA was also predicted as antileishmanial molecule corroborating the experimental activity. Therefore, HA is a promising antileishmanial molecule worthy of further development as a biotherapeutic agent.
Collapse
Affiliation(s)
- Justice Afrifa Crentsil
- Department of Chemistry, School of Physical and Mathematical Sciences, College of Basic and Applied Sciences (CBAS), University of Ghana, Accra, Ghana
| | - Lauve Rachel Tchokouaha Yamthe
- Institute for Medical Research and Medicinal Plants Studies, Yaoundé, Cameroon.,Department of Parasitology, College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana.,Antimicrobial and Biocontrol Agents Unit, Laboratory for Phytobiochemistry and Medicinal Plants Studies, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon
| | - Barbara Zenabu Anibea
- Department of Chemistry, School of Physical and Mathematical Sciences, College of Basic and Applied Sciences (CBAS), University of Ghana, Accra, Ghana
| | - Emmanuel Broni
- Department of Biomedical Engineering, School of Engineering Sciences, CBAS, University of Ghana, Accra, Ghana
| | - Samuel Kojo Kwofie
- Department of Biomedical Engineering, School of Engineering Sciences, CBAS, University of Ghana, Accra, Ghana.,West African Centre for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, CBAS, University of Ghana, Accra, Ghana.,Department of Medicine, Loyola University Medical Center, Maywood, IL, United States.,Department of Physics and Engineering Science, Coastal Carolina University, Conway, SC, United States
| | - John Kweku Amissah Tetteh
- Department of Immunology, College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Dorcas Osei-Safo
- Department of Chemistry, School of Physical and Mathematical Sciences, College of Basic and Applied Sciences (CBAS), University of Ghana, Accra, Ghana
| |
Collapse
|
30
|
Ranatunga M, Rai R, Richardson SCW, Dyer P, Harbige L, Deacon A, Pecorino L, Getti GTM. Leishmania aethiopica cell-to-cell spreading involves caspase-3, AkT, and NF-κB but not PKC-δ activation and involves uptake of LAMP-1-positive bodies containing parasites. FEBS J 2020; 287:1777-1797. [PMID: 31804757 DOI: 10.1111/febs.15166] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 08/27/2019] [Accepted: 12/04/2019] [Indexed: 11/25/2022]
Abstract
Development of human leishmaniasis is dependent on the ability of intracellular Leishmania parasites to spread and enter macrophages. The mechanism through which free promastigotes and amastigotes bind and enter host macrophages has been previously investigated; however, little is known about intracellular trafficking and cell-to-cell spreading. In this study, the mechanism involved in the spreading of Leishmania aethiopica and Leishmania mexicana was investigated. A significant increase in phosphatidylserine (PS) exhibition, cytochrome C release, and active caspase-3 expression was detected (P < 0.05) during L. aethiopica, but not L. mexicana spreading. A decrease (P < 0.05) of protein kinase B (Akt) protein and BCL2-associated agonist of cell death (BAD) phosphorylation was also observed. The nuclear factor kappa-light-chain enhancer of activated B cells (NF-kB) signaling pathway and pro-apoptotic protein protein kinase C delta (PKC-δ) were downregulated while inhibition of caspase-3 activation prevented L. aethiopica spreading. Overall suggesting that L. aethiopica induces host cell's apoptosis during spreading in a caspase-3-dependent manner. The trafficking of amastigotes within macrophages following cell-to-cell spreading differed from that of axenic parasites and involved co-localization with lysosomal-associated membrane protein 1 (LAMP-1) within 10 min postinfection. Interestingly, following infection with axenic amastigotes and promastigotes, co-localization of parasites with LAMP-1-positive structures took place at 1 and 4 h, respectively, suggesting that the membrane coat and LAMP-1 protein were derived from the donor cell. Collectively, these findings indicate that host cell apoptosis, demonstrated by PS exhibition, caspase-3 activation, cytochrome C release, downregulation of Akt, BAD phosphorylation, NF-kB activation, and independent of PKC-δ expression, is involved in L. aethiopica spreading. Moreover, L. aethiopica parasites associate with LAMP-rich structures when taken up by neighboring macrophages.
Collapse
Affiliation(s)
| | - Rajeev Rai
- University of Greenwich at Medway, Kent, UK
| | | | - Paul Dyer
- University of Greenwich at Medway, Kent, UK
| | | | | | | | | |
Collapse
|
31
|
Vivarini ADC, Lopes UG. The Potential Role of Nrf2 Signaling in Leishmania Infection Outcomes. Front Cell Infect Microbiol 2020; 9:453. [PMID: 31998662 PMCID: PMC6966304 DOI: 10.3389/fcimb.2019.00453] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 12/13/2019] [Indexed: 01/06/2023] Open
Abstract
Nrf2 [nuclear factor erythroid 2-related factor 2 (Nrf2)] regulates the expression of a plethora of genes involved in the response to oxidative stress due to inflammation, aging, and tissue damage, among other pathological conditions. Deregulation of this cytoprotective system may also interfere with innate and adaptive immune responses. Oxidative burst, one of the main microbicidal mechanisms, could be impaired during initial phagocytosis of parasites, which could lead to the successful establishment of infection and promote susceptibility to diseases. A wide diversity of infections, mainly those caused by intracellular pathogens such as viruses, bacteria, and protozoan parasites, modulate the activation of Nrf2 by interfering with post-translational modifications, interactions between different protein complexes and the immune response. Nrf2 may be induced by pathogens via distinct pathways such as those involving the engagement of Toll-like receptors, the activation of PI3K/Akt, and endoplasmic reticulum stress. Recent studies have revealed the importance of Nrf2 on leishmaniasis. This mini-review discusses relevant findings that reveal the connection between Leishmania-induced modifications of the host pathways and their relevance to the modulation of the Nrf2-dependent antioxidative response to the infection.
Collapse
Affiliation(s)
- Aislan de Carvalho Vivarini
- Laboratory of Molecular Parasitology, Center of Health Science, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ulisses Gazos Lopes
- Laboratory of Molecular Parasitology, Center of Health Science, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
32
|
Chandrakar P, Parmar N, Descoteaux A, Kar S. Differential Induction of SOCS Isoforms by Leishmania donovani Impairs Macrophage–T Cell Cross-Talk and Host Defense. THE JOURNAL OF IMMUNOLOGY 2019; 204:596-610. [DOI: 10.4049/jimmunol.1900412] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 11/24/2019] [Indexed: 12/31/2022]
|
33
|
Chen R, Ji G, Ren H, Liu Z, Feng S, Zhang T, Xi L, Li X. Activation of autophagy and IL-10 production are regulated by Jun N-terminal kinase 1 and 2 and p38 mitogen activated protein kinase signaling pathways during Talaromyces marneffei infection within dendritic cells. Microb Pathog 2019; 139:103891. [PMID: 31783123 DOI: 10.1016/j.micpath.2019.103891] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 11/01/2019] [Accepted: 11/23/2019] [Indexed: 11/16/2022]
Abstract
Previous study have shown that Talaromyces marneffei (T. marneffei) induced activation of autophagy. Therefore, we explore signaling pathway that regulates activation of autophagy by intracellular signaling mechanisms during T. marneffei infection. Further, we examine c-Jun N-terminal kinase 1 and 2 (JNK1/2) and p38 signaling pathways that regulate IL-1β and IL-10 production and activation of autophagy during T. marneffei infection in human dendritic cells (DCs). We found that T. marneffei induced activation of JNK1/2 and p38 in human DCs. Furthermore, the inhibition of JNK1/2 and p38 increased activation of autophagy and decreased the replication of T. marneffei in T. marneffei-infected human DCs. Moreover, IL-1β secretion in T. marneffei-infected human DCs was dependent on JNK1/2 and autophagy pathways, whereas IL-10 secretion was dependent on JNK1/2, p38 and autophagy pathways. These data suggest that JNK1/2 and p38 pathways play critical roles in activation of autophagy, the multiplication of T. marneffei and subsequent cytokine production during T. marneffei infection.
Collapse
Affiliation(s)
- Renqiong Chen
- Department of Dermatology, Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, 222002, China; Department of Dermatology, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang, 222002, China.
| | - Guangquan Ji
- Department of Technology, Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, 222002, China
| | - Hong Ren
- Department of Dermatology, Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, 222002, China; Department of Dermatology, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang, 222002, China.
| | - Zhonglun Liu
- Department of Dermatology, Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, 222002, China; Department of Dermatology, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang, 222002, China
| | - Shan Feng
- Department of Technology, Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, 222002, China
| | - Tingting Zhang
- Department of Infection, Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, 222002, China
| | - Liyan Xi
- Department of Dermatology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
| | - Xiaoming Li
- Department of Emergency, Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, 222002, China.
| |
Collapse
|
34
|
Gervais O, Chollet B, Dubreuil C, Durante S, Feng C, Hénard C, Lecadet C, Serpin D, Tristan R, Arzul I. Involvement of apoptosis in the dialogue between the parasite Bonamia ostreae and the flat oyster Ostrea edulis. FISH & SHELLFISH IMMUNOLOGY 2019; 93:958-964. [PMID: 31442589 DOI: 10.1016/j.fsi.2019.08.035] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 07/05/2019] [Accepted: 08/14/2019] [Indexed: 06/10/2023]
Abstract
The protozoan parasite Bonamia ostreae has been associated with the decline of flat oyster Ostrea edulis populations in some European countries. Control of shellfish diseases mostly relies on prevention measures including transfer restrictions and stock management measures such as breeding programmes. These prevention and mitigation measures require a better understanding of interactions between host and pathogens. Previous in vitro studies allowed identifying apoptosis as a mechanism activated by the flat oyster in response to B. ostreae. However, these experiments also suggested that the parasite is able to regulate apoptosis in order to survive and multiply within hemocytes. By simplifying the conditions of infection, in vitro studies allow identifying most distinct features of the response of the host. In order to appreciate the relative importance of apoptosis in this response at the oyster scale, in vivo trials were carried out by injecting with parasites oysters from two French locations, Quiberon Bay (Brittany) and Diana Lagoon (Corsica). Apoptosis was investigated on pools of hemolymph from oysters collected at early and later times after injection using previously developed tools. Apoptotic cellular activities including intracytoplasmic calcium concentration, mitochondrial membrane potential and phosphatidyl serine externalization were analysed using flow cytometry. Moreover, the expression of flat oyster genes involved in both extrinsic and intrinsic pathways was measured using real time quantitative PCR.
Collapse
Affiliation(s)
- Ophélie Gervais
- Ifremer, RBE-SG2M-LGPMM, Station de La Tremblade, Avenue de Mus de Loup, F-17390, La Tremblade, France
| | - Bruno Chollet
- Ifremer, RBE-SG2M-LGPMM, Station de La Tremblade, Avenue de Mus de Loup, F-17390, La Tremblade, France
| | - Christine Dubreuil
- Ifremer, RBE-SG2M-LGPMM, Station de La Tremblade, Avenue de Mus de Loup, F-17390, La Tremblade, France
| | - Serena Durante
- Università veterinaria di Milano, Via Giovanni Celoria, 20133, Milano, Italy
| | - Chunyan Feng
- Institute of Animal Quarantine Chinese Academy of Inspection, Beijing, China
| | - Cyril Hénard
- Ifremer, RBE-SG2M-LGPMM, Station de La Tremblade, Avenue de Mus de Loup, F-17390, La Tremblade, France
| | - Cyrielle Lecadet
- Ifremer, RBE-SG2M-LGPMM, Station de La Tremblade, Avenue de Mus de Loup, F-17390, La Tremblade, France
| | - Delphine Serpin
- Ifremer, RBE-SG2M-LGPMM, Station de La Tremblade, Avenue de Mus de Loup, F-17390, La Tremblade, France
| | - Renault Tristan
- Ifremer, RBE, Centre de Nantes, Rue de l'Ile d'Yeu, F-44311, Nantes, France
| | - Isabelle Arzul
- Ifremer, RBE-SG2M-LGPMM, Station de La Tremblade, Avenue de Mus de Loup, F-17390, La Tremblade, France.
| |
Collapse
|
35
|
Zahid MSH, Johnson MM, Tokarski RJ, Satoskar AR, Fuchs JR, Bachelder EM, Ainslie KM. Evaluation of synergy between host and pathogen-directed therapies against intracellular Leishmania donovani. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2019; 10:125-132. [PMID: 31493763 PMCID: PMC6731340 DOI: 10.1016/j.ijpddr.2019.08.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 08/16/2019] [Accepted: 08/21/2019] [Indexed: 11/24/2022]
Abstract
Visceral leishmaniasis (VL) is associated with treatment complications due to the continued growth of resistant parasites toward currently available pathogen-directed therapeutics. To limit the emergence and combat resistant parasites there is a need to develop new anti-leishmanial drugs and alternative treatment approaches, such as host-directed therapeutics (HDTs). Discovery of new anti-leishmanial drugs including HDTs requires suitable in vitro assay systems. Herein, we modified and evaluated a series of resazurin assays against different life-stages of the VL causing parasite, Leishmania donovani to identify novel HDTs. We further analyzed the synergy of combinatorial interactions between traditionally used pathogen-directed drugs and HDTs for clearance of intracellular L. donovani. The inhibitory concentration at 50% (IC50) of the five evaluated therapies [amphotericin B (AMB), miltefosine, paromomycin, DNER-4, and AR-12 (OSU-03012)] was determined against promastigotes, extracellular amastigotes, and intracellular amastigotes of L. donovani via a resazurin-based assay and compared to image-based microscopy. Using the resazurin-based assay, all evaluated therapies showed reproducible anti-leishmanial activity against the parasite's different life-stages. These results were consistent to the traditional image-based technique. The gold standard of therapy, AMB, showed the highest potency against intracellular L. donovani, and was further evaluated for combinatorial effects with the HDTs. Among the combinations analyzed, pathogen-directed AMB and host-directed AR-12 showed a synergistic reduction of intracellular L. donovani compared to individual treatments. The modified resazurin assay used in this study demonstrated a useful technique to measure new anti-leishmanial drugs against both intracellular and extracellular parasites. The synergistic interactions between pathogen-directed AMB and host-directed AR-12 showed a great promise to combat VL, with the potential to reduce the emergence of drug-resistant strains.
Collapse
Affiliation(s)
- M Shamim Hasan Zahid
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Monica M Johnson
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Robert J Tokarski
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Abhay R Satoskar
- Department of Pathology, Medical Center, The Ohio State University, Columbus, OH, 43210, USA
| | - James R Fuchs
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Eric M Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Kristy M Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
36
|
Borbón TY, Scorza BM, Clay GM, Lima Nobre de Queiroz F, Sariol AJ, Bowen JL, Chen Y, Zhanbolat B, Parlet CP, Valadares DG, Cassel SL, Nauseef WM, Horswill AR, Sutterwala FS, Wilson ME. Coinfection with Leishmania major and Staphylococcus aureus enhances the pathologic responses to both microbes through a pathway involving IL-17A. PLoS Negl Trop Dis 2019; 13:e0007247. [PMID: 31107882 PMCID: PMC6527190 DOI: 10.1371/journal.pntd.0007247] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 02/15/2019] [Indexed: 12/12/2022] Open
Abstract
Cutaneous leishmaniasis (CL) is a parasitic disease causing chronic, ulcerating skin lesions. Most humans infected with the causative Leishmania protozoa are asymptomatic. Leishmania spp. are usually introduced by sand flies into the dermis of mammalian hosts in the presence of bacteria from either the host skin, sand fly gut or both. We hypothesized that bacteria at the dermal inoculation site of Leishmania major will influence the severity of infection that ensues. A C57BL/6 mouse ear model of single or coinfection with Leishmania major, Staphylococcus aureus, or both showed that single pathogen infections caused localized lesions that peaked after 2–3 days for S. aureus and 3 weeks for L. major infection, but that coinfection produced lesions that were two-fold larger than single infection throughout 4 weeks after coinfection. Coinfection increased S. aureus burdens over 7 days, whereas L. major burdens (3, 7, 28 days) were the same in singly and coinfected ears. Inflammatory lesions throughout the first 4 weeks of coinfection had more neutrophils than did singly infected lesions, and the recruited neutrophils from early (day 1) lesions had similar phagocytic and NADPH oxidase capacities. However, most neutrophils were apoptotic, and transcription of immunomodulatory genes that promote efferocytosis was not upregulated, suggesting that the increased numbers of neutrophils may, in part, reflect defective clearance and resolution of the inflammatory response. In addition, the presence of more IL-17A-producing γδ and non-γδ T cells in early lesions (1–7 days), and L. major antigen-responsive Th17 cells after 28 days of coinfection, with a corresponding increase in IL-1β, may recruit more naïve neutrophils into the inflammatory site. Neutralization studies suggest that IL-17A contributed to an enhanced inflammatory response, whereas IL-1β has an important role in controlling bacterial replication. Taken together, these data suggest that coinfection of L. major infection with S. aureus exacerbates disease, both by promoting more inflammation and neutrophil recruitment and by increasing neutrophil apoptosis and delaying resolution of the inflammatory response. These data illustrate the profound impact that coinfecting microorganisms can exert on inflammatory lesion pathology and host adaptive immune responses. Cutaneous leishmaniasis (CL) is a vector-borne ulcerating skin disease affecting several million people worldwide. The causative Leishmania spp. protozoa are transmitted by infected phlebotomine sand flies. During a sand fly bite, bacteria can be coincidentally inoculated into the dermis with the parasite. Staphylococcus aureus is the most common bacterium in CL skin lesions. Symptomatic CL is characterized by papulonodular skin lesions that ulcerate and resolve with scarring, although most cutaneous Leishmania infections are asymptomatic. We sought to explore factors that determine whether infection with a cutaneous Leishmania species would result in symptomatic CL rather than asymptomatic infection. We hypothesized that local bacteria promote the development of symptomatic CL lesions during infection with Leishmania major. We discovered that cutaneous lesions were significantly larger in mice inoculated simultaneously with S. aureus and L. major than in mice infected with either organism alone. Coinfection led to increased S. aureus growth in skin lesions, whereas L. major parasite numbers were unchanged by coinfection. The size of the exacerbated lesion correlated with early increased numbers of neutrophils and elevated levels of proinflammatory cytokines IL-1β and IL-17A during the first 7 days, and with sustained increases in IL-17A through 28 days of coinfection. Neutralizing antibody experiments suggested IL-17A was partially responsible for lesion exacerbation during coinfection, whereas IL-1β was important for both control of early lesion exacerbation and promotion of IL-17A production. These data suggest that treatment of symptomatic CL targeting the parasite, local commensal bacteria, and host proinflammatory IL-17A immune responses might improve the outcome of CL.
Collapse
Affiliation(s)
- Tiffany Y. Borbón
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States of America
- Medical Scientist Training Program and the Carver College of Medicine, University of Iowa, Iowa City, IA, United States of America
| | - Breanna M. Scorza
- Interdisciplinary Ph.D. Program in Immunology, University of Iowa, Iowa City, IA, United States of America
| | - Gwendolyn M. Clay
- Medical Scientist Training Program and the Carver College of Medicine, University of Iowa, Iowa City, IA, United States of America
- Interdisciplinary Ph.D. Program in Molecular Medicine, University of Iowa, Iowa City, IA, United States of America
| | | | - Alan J. Sariol
- Interdisciplinary Ph.D. Program in Immunology, University of Iowa, Iowa City, IA, United States of America
| | - Jayden L. Bowen
- Medical Scientist Training Program and the Carver College of Medicine, University of Iowa, Iowa City, IA, United States of America
| | - Yani Chen
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa City, IA, United States of America
- Iowa Inflammation Program, Department of Internal Medicine, University of Iowa, Iowa City, IA, United States of America
- Veterans’ Affairs Medical Center, Iowa City, IA, United States of America
| | - Bayan Zhanbolat
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa City, IA, United States of America
- Iowa Inflammation Program, Department of Internal Medicine, University of Iowa, Iowa City, IA, United States of America
- Veterans’ Affairs Medical Center, Iowa City, IA, United States of America
| | - Corey P. Parlet
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States of America
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa City, IA, United States of America
- Iowa Inflammation Program, Department of Internal Medicine, University of Iowa, Iowa City, IA, United States of America
| | - Diogo G. Valadares
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States of America
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa City, IA, United States of America
- Iowa Inflammation Program, Department of Internal Medicine, University of Iowa, Iowa City, IA, United States of America
- Veterans’ Affairs Medical Center, Iowa City, IA, United States of America
- Conselho Nacional de Desenvolvimento Cientifico e Tecnológico (CNPq), Brasilia, Brazil
| | - Suzanne L. Cassel
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States of America
| | - William M. Nauseef
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States of America
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa City, IA, United States of America
- Iowa Inflammation Program, Department of Internal Medicine, University of Iowa, Iowa City, IA, United States of America
- Veterans’ Affairs Medical Center, Iowa City, IA, United States of America
| | - Alexander R. Horswill
- Department of Immunology and Microbiology, University of Colorado Denver—Anschutz Medical Campus, Aurora, CO, United States of America
| | - Fayyaz S. Sutterwala
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States of America
| | - Mary E. Wilson
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States of America
- Medical Scientist Training Program and the Carver College of Medicine, University of Iowa, Iowa City, IA, United States of America
- Interdisciplinary Ph.D. Program in Immunology, University of Iowa, Iowa City, IA, United States of America
- Interdisciplinary Ph.D. Program in Molecular Medicine, University of Iowa, Iowa City, IA, United States of America
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa City, IA, United States of America
- Iowa Inflammation Program, Department of Internal Medicine, University of Iowa, Iowa City, IA, United States of America
- Veterans’ Affairs Medical Center, Iowa City, IA, United States of America
- * E-mail:
| |
Collapse
|
37
|
Roy S, Saha S, Gupta P, Ukil A, Das PK. Crosstalk of PD-1 signaling with the SIRT1/FOXO-1 axis during the progression of visceral leishmaniasis. J Cell Sci 2019; 132:jcs.226274. [PMID: 30910830 DOI: 10.1242/jcs.226274] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 03/20/2019] [Indexed: 12/16/2022] Open
Abstract
Previously, we documented the role of the programmed death-1 (PD-1, also known as PDCD1) pathway in macrophage apoptosis and the downregulation of this signaling during infection by the intra-macrophage parasite Leishmania donovani However, we also found that, during the late phase of infection, PD-1 expression was significantly increased without activating host cell apoptosis; here we show that inhibition of PD-1 led to markedly decreased parasite survival, along with increased production of TNFα, IL-12, reactive oxygen species (ROS) and nitric oxide (NO). Increased PD-1 led to inactivation of AKT proteins resulting in nuclear sequestration of FOXO-1. Transfecting infected cells with constitutively active FOXO-1 (CA-FOXO) led to increased cell death, thereby suggesting that nuclear FOXO-1 might be inactivated. Infection significantly induced the expression of SIRT1, which inactivated FOXO-1 through deacetylation, and its knockdown led to increased apoptosis. SIRT1 knockdown also significantly decreased parasite survival along with increased production of TNFα, ROS and NO. Administration of the SIRT1 inhibitor sirtinol (10 mg/kg body weight) in infected mice decreased spleen parasite burden and a synergistic effect was found with PD-1 inhibitor. Collectively, our study shows that Leishmania utilizes the SIRT1/FOXO-1 axis for differentially regulating PD-1 signaling and, although they are interconnected, both pathways independently contribute to intracellular parasite survival.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Shalini Roy
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Shriya Saha
- Department of Biochemistry, Calcutta University, Kolkata 700019, India
| | - Purnima Gupta
- Department of Biochemistry, Calcutta University, Kolkata 700019, India
| | - Anindita Ukil
- Department of Biochemistry, Calcutta University, Kolkata 700019, India
| | - Pijush K Das
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| |
Collapse
|
38
|
Laha B, Verma AK, Biswas B, Sengodan SK, Rastogi A, Willard B, Ghosh M. Detection and characterization of an albumin-like protein in Leishmania donovani. Parasitol Res 2019; 118:1609-1623. [PMID: 30903348 DOI: 10.1007/s00436-019-06286-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 03/04/2019] [Indexed: 12/17/2022]
Abstract
The protozoan parasite, Leishmania donovani, undergoes several molecular adaptations and secretes many effector molecules for host cell manipulation and successful parasitism. The current study identifies an albumin-like secretory protein, expressed in its extracellular promastigote forms. A leishmanial complementary DNA sequence of a partial gene has been cloned, and the encoded peptide (14 kD) is used for the production of polyclonal antibody. This targeted antibody identifies a large native protein (66.421 kD), expressed stage-specifically in promastigotes. Through electron microscopic studies, the native protein is found to be localized in the flagellar pocket and flagella and at the surface of the promastigotes. This native protein is purified with the same customized antibody for future characterization and sequencing. The sequence analysis reveals its homology with the mammalian serum albumin. It is evidenced from in silico studies that this albumin-like protein remains associated with long-chain fatty acids while in vitro studies indicate its close association with membrane cholesterol. Since antibody-mediated blocking compromises the parasite infectivity, these leishmanial albumin-like molecules are hereby proposed to play an instrumental role in the infectivity of L. donovani to peripheral blood monocyte cells. Thus, identification and characterization of an albumin-like protein in L. donovani promastigotes may be interpreted as a molecular adaptation candidate. It may be hypothesized that the parasite mimics the mammalian system for importing fatty acids into the intracellular amastigotes, facilitating its host cell infectivity.
Collapse
Affiliation(s)
- Bhakti Laha
- Department of Biotechnology, National Institute of Technology Durgapur, Mahatma Gandhi Avenue, Durgapur, West Bengal, 713209, India
| | - Amit Kumar Verma
- Department of Biotechnology, National Institute of Technology Durgapur, Mahatma Gandhi Avenue, Durgapur, West Bengal, 713209, India
| | - Bapi Biswas
- Department of Biotechnology, National Institute of Technology Durgapur, Mahatma Gandhi Avenue, Durgapur, West Bengal, 713209, India
| | - Satheesh Kumar Sengodan
- Department of Biotechnology, National Institute of Technology Durgapur, Mahatma Gandhi Avenue, Durgapur, West Bengal, 713209, India
| | - Akanksha Rastogi
- Department of Biotechnology, National Institute of Technology Durgapur, Mahatma Gandhi Avenue, Durgapur, West Bengal, 713209, India
| | - Belinda Willard
- Mass Spectrometry Laboratory for Protein Sequencing, Cleveland Clinic - Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Monidipa Ghosh
- Department of Biotechnology, National Institute of Technology Durgapur, Mahatma Gandhi Avenue, Durgapur, West Bengal, 713209, India.
| |
Collapse
|
39
|
Encephalitozoon cuniculi and Vittaforma corneae (Phylum Microsporidia) inhibit staurosporine-induced apoptosis in human THP-1 macrophages in vitro. Parasitology 2018; 146:569-579. [PMID: 30486909 DOI: 10.1017/s0031182018001968] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Obligately intracellular microsporidia regulate their host cell life cycles, including apoptosis, but this has not been evaluated in phagocytic host cells such as macrophages that can facilitate infection but also can be activated to kill microsporidia. We examined two biologically dissimilar human-infecting microsporidia species, Encephalitozoon cuniculi and Vittaforma corneae, for their effects on staurosporine-induced apoptosis in the human macrophage-differentiated cell line, THP1. Apoptosis was measured after exposure of THP-1 cells to live and dead mature organisms via direct fluorometric measurement of Caspase 3, colorimetric and fluorometric TUNEL assays, and mRNA gene expression profiles using Apoptosis RT2 Profiler PCR Array. Both species of microsporidia modulated the intrinsic apoptosis pathway. In particular, live E. cuniculi spores inhibited staurosporine-induced apoptosis as well as suppressed pro-apoptosis genes and upregulated anti-apoptosis genes more broadly than V. corneae. Exposure to dead spores induced an opposite effect. Vittaforma corneae, however, also induced inflammasome activation via Caspases 1 and 4. Of the 84 apoptosis-related genes assayed, 42 (i.e. 23 pro-apoptosis, nine anti-apoptosis, and 10 regulatory) genes were more affected including those encoding members of the Bcl2 family, caspases and their regulators, and members of the tumour necrosis factor (TNF)/TNF receptor R superfamily.
Collapse
|
40
|
Nandan D, Zhang N, Yu Y, Schwartz B, Chen S, Kima PE, Reiner NE. Miransertib (ARQ 092), an orally-available, selective Akt inhibitor is effective against Leishmania. PLoS One 2018; 13:e0206920. [PMID: 30399177 PMCID: PMC6219794 DOI: 10.1371/journal.pone.0206920] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 10/22/2018] [Indexed: 12/25/2022] Open
Abstract
Leishmaniasis is amongst the most important neglected diseases, afflicting more than 12 million people in 88 countries. There is an urgent need for safe orally bioavailable and cost-effective drugs for the treatment of leishmaniasis. It has recently been shown that Leishmania activates host macrophage serine/threonine kinase Akt, to promote survival of both parasites and infected cells. Here, we sought to evaluate a compound, Miransertib (ARQ 092), an orally bioavailable and selective allosteric Akt inhibitor currently in clinical trials for patients with PI3K/Akt-driven tumors or Proteus syndrome. Miransertib was tested against Leishmania donovani and Leishmania amazonensis, causative agents of visceral and cutaneous leishmaniasis, respectively. Cultured promastigotes were susceptible to Miransertib. In addition, Miransertib was markedly effective against intracellular amastigotes of L. donovani or L. amazonensis-infected macrophages. Miransertib also enhanced mTOR dependent autophagy in Leishmania-infected macrophages, which may represent one mechanism of Miransertib-mediated killing of intracellular Leishmania. Whereas parasite clearance in the spleen of mice infected with L. donovani and treated with Miransertib was comparable to that when treated with miltefosine, Miransertib caused a greater reduction in the parasite load in the liver. In the cutaneous leishmaniasis infection model, lesions were reduced by 40% as compared to mock treated mice. Together, these results provide direct evidence to support the conclusion that Miransertib is an excellent lead compound for the development of a new oral drug therapy for visceral and cutaneous leishmaniasis.
Collapse
Affiliation(s)
- Devki Nandan
- Department of Medicine, Division of Infectious Diseases, University of British Columbia, Vancouver, BC, Canada
| | - Naixin Zhang
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, United States of America
| | - Yi Yu
- ArQule, Inc, Burlington, Massachusetts, United States of America
| | - Brian Schwartz
- ArQule, Inc, Burlington, Massachusetts, United States of America
| | - Stella Chen
- Department of Medicine, Division of Infectious Diseases, University of British Columbia, Vancouver, BC, Canada
| | - Peter E. Kima
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, United States of America
| | - Neil E. Reiner
- Department of Medicine, Division of Infectious Diseases, University of British Columbia, Vancouver, BC, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
- * E-mail:
| |
Collapse
|
41
|
Sousa R, Andrade VM, Bair T, Ettinger NA, Guimarães L, Andrade L, Guimarães LH, Machado PRL, Carvalho EM, Wilson ME, Schriefer A. Early Suppression of Macrophage Gene Expression by Leishmania braziliensis. Front Microbiol 2018; 9:2464. [PMID: 30374342 PMCID: PMC6196312 DOI: 10.3389/fmicb.2018.02464] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 09/26/2018] [Indexed: 01/27/2023] Open
Abstract
Leishmania braziliensis is an intracellular parasite that resides mostly in macrophages. Both the parasite genome and the clinical disease manifestations show considerable polymorphism. Clinical syndromes caused by L. braziliensis include localized cutaneous (CL), mucosal (ML), and disseminated leishmaniasis (DL). Our prior studies showed that genetically distinct L. braziliensis clades associate with different clinical types. Herein, we hypothesized that: (1) L. braziliensis induces changes in macrophage gene expression that facilitates infection; (2) infection of macrophages with strains associated with CL (clade B), ML (clade C), or DL (clade A) will differentially affect host cell gene expression, reflecting their different pathogenic mechanisms; and (3) differences between the strains will be reflected by differences in macrophage gene expression after initial exposure to the parasite. Human monocyte derived macrophages were infected with L. braziliensis isolates from clades A, B, or C. Patterns of gene expression were compared using Affymetrix DNA microarrays. Many transcripts were significantly decreased by infection with all isolates. The most dramatically decreased transcripts encoded proteins involved in signaling pathways, apoptosis, or mitochondrial oxidative phosphorylation. Some transcripts encoding stress response proteins were up-regulated. Differences between L. braziliensis clades were observed in the magnitude of change, rather than the identity of transcripts. Isolates from subjects with metastatic disease (ML and DL) induced a greater magnitude of change than isolates from CL. We conclude that L. braziliensis enhances its intracellular survival by inhibiting macrophage pathways leading to microbicidal activity. Parasite strains destined for dissemination may exert a more profound suppression than less invasive L. braziliensis strains that remain near the cutaneous site of inoculation.
Collapse
Affiliation(s)
- Rosana Sousa
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | - Viviane M Andrade
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | - Thomas Bair
- DNA Facility, The University of Iowa, Iowa City, IA, United States
| | - Nicholas A Ettinger
- Deptartment of Pediatrics-Critical Care, Baylor College of Medicine, Houston, TX, United States
| | - Luana Guimarães
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | - Laura Andrade
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | - Luiz H Guimarães
- Centro de Formação em Saúde, Universidade Federal do Sul da Bahia, Teixeira de Freitas, Brazil.,Instituto Nacional de Ciência e Tecnologia - Doenças Tropicais, Salvador, Brazil
| | - Paulo R L Machado
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil.,Instituto Nacional de Ciência e Tecnologia - Doenças Tropicais, Salvador, Brazil
| | - Edgar M Carvalho
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil.,Instituto Nacional de Ciência e Tecnologia - Doenças Tropicais, Salvador, Brazil.,Instituto Gonçalo Moniz, FIOCRUZ, Salvador, Brazil
| | - Mary E Wilson
- Departments of Internal Medicine and Microbiology, VA Medical Center, The University of Iowa, Iowa City, IA, United States
| | - Albert Schriefer
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil.,Instituto Nacional de Ciência e Tecnologia - Doenças Tropicais, Salvador, Brazil.,Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| |
Collapse
|
42
|
Molecular and cellular characterization of apoptosis in flat oyster a key mechanisms at the heart of host-parasite interactions. Sci Rep 2018; 8:12494. [PMID: 30131502 PMCID: PMC6104086 DOI: 10.1038/s41598-018-29776-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 07/14/2018] [Indexed: 01/09/2023] Open
Abstract
Bonamia ostreae has been associated with the decline of flat oyster Ostrea edulis populations in some European countries. This obligatory intracellular parasite persists and multiplies into hemocytes. Previous in vitro experiments showed that apoptosis is activated in hemocytes between 1 h and 4 h of contact with the parasite. The flat oyster uses the apoptosis pathway to defend against B. ostreae. However, the parasite might be also able to modulate this response in order to survive in its host. In order to investigate this hypothesis the apoptotic response of the host was evaluated using flow cytometry, transmission electron microscopy and by measuring the response of genes involved in the apoptotic pathway after 4 h. In parallel, the parasite response was investigated by measuring the expression of B. ostreae genes involved in different biological functions including cell cycle and cell death. Obtained results allow describing molecular apoptotic pathways in O. edulis and confirm that apoptosis is early activated in hemocytes after a contact with B. ostreae. Interestingly, at cellular and molecular levels this process appeared downregulated after 44 h of contact. Concurrently, parasite gene expression appeared reduced suggesting that the parasite could inhibit its own metabolism to escape the immune response.
Collapse
|
43
|
Zhang N, Prasad S, Huyghues Despointes CE, Young J, Kima PE. Leishmania parasitophorous vacuole membranes display phosphoinositides that create conditions for continuous Akt activation and a target for miltefosine in Leishmania infections. Cell Microbiol 2018; 20:e12889. [PMID: 29993167 DOI: 10.1111/cmi.12889] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 06/22/2018] [Accepted: 06/25/2018] [Indexed: 12/13/2022]
Abstract
Miltefosine is an important drug for the treatment of leishmaniasis; however, its mechanism of action is still poorly understood. In these studies, we tested the hypothesis that like in cancer cells, miltefosine's efficacy in leishmaniasis is due to its inhibition of Akt activation in host cells. We show using pharmacologic agents that block Akt activation by different mechanisms and also using an inducible knockdown approach that miltefosine loses its efficacy when its access to Akt1 is limited. Interestingly, limitation of Akt activation results in clearance of established Leishmania infections. We then show, using fluorophore-tagged probes that bind to phosphoinositides, that Leishmania parasitophorous vacuole membranes (LPVMs) display the relevant phosphoinositides to which Akt can be recruited and activated continuously. Taken together, we propose that the acquisition of PI(4) P and the display of PI (3,4)P2 on LPVMs initiate the machinery that supports continuous Akt activation and sensitivity to miltefosine.
Collapse
Affiliation(s)
- Naixin Zhang
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, USA
| | - Samiksha Prasad
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, USA
| | | | - Jeffrey Young
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, USA
| | - Peter E Kima
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
44
|
Abhishek K, Das S, Kumar A, Kumar A, Kumar V, Saini S, Mandal A, Verma S, Kumar M, Das P. Leishmania donovani induced Unfolded Protein Response delays host cell apoptosis in PERK dependent manner. PLoS Negl Trop Dis 2018; 12:e0006646. [PMID: 30036391 PMCID: PMC6081962 DOI: 10.1371/journal.pntd.0006646] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 08/02/2018] [Accepted: 06/29/2018] [Indexed: 02/06/2023] Open
Abstract
Background Endoplasmic reticulum (ER) stress generated unfolded stress response (UPR) is a basic survival mechanism which protects cell under unfavourable conditions. Leishmania parasite modulates host macrophages in various ways to ensure its survival. Modulation of PI3K-Akt pathway in delayed apoptotic induction of host; enables parasite to stabilize the infection for further propagation. Methodology Infected RAW macrophages were exposed to campothecin or thagsigargin and phosphorylation status of PERK, Akt, BAD and Cyt-C was determined through western blotting using phospho specific antibody. Expression at transcriptional level for cIAP1 &2, ATF4, CHOP, ATF3, HO-1 and sXBP1 was determined using real time PCR. For inhibition studies, RAW macrophages were pre-treated with PERK inhibitor GSK2606414 before infection. Findings Our studies in RAW macrophages showed that induction of host UPR against L.donovani infection activates Akt mediated pathway which delays apoptotic induction of the host. Moreover, Leishmania infection results in phosphorylation and activation of host PERK enzyme and increased transcription of genes of inhibitor of apoptosis gene family (cIAP) mRNA. In our inhibition studies, we found that inhibition of infection induced PERK phosphorylation under apoptotic inducers reduces the Akt phosphorylation and fails to activate further downstream molecules involved in protection against apoptosis. Also, inhibition of PERK phosphorylation under oxidative exposure leads to increased Nitric Oxide production. Simultaneously, decreased transcription of cIAP mRNA upon PERK phosphorylation fates the host cell towards apoptosis hence decreased infection rate. Conclusion Overall the findings from the study suggests that Leishmania modulated host UPR and PERK phosphorylation delays apoptotic induction in host macrophage, hence supports parasite invasion at early stages of infection. Visceral Leishmaniasis or Kala-azar is one of the severe tropical neglected parasitic diseases caused by Leishmania donovani in Indian subcontinent. Modulation of host in terms of delayed apoptotic induction is one of the aspects which favours disease establishment; however the mechanism is not clearly understood yet. In the present study, we tried to explore the connection between L.donovani infection induced UPR in host with delayed onset of apoptosis. We found that L.donovani infection phosphorylates the PERK and Akt molecule in host along with delayed apoptosis. Simultaneously, the levels of cellular IAP (cIAP1 & 2) genes were also up-regulated in infected macrophages. To assess the involvement of PERK in delayed apoptosis of host, we inhibited the phosphorylation of PERK under the exposure to apoptotic inducers. We found that PERK inhibition decreased the Akt phosphorylation and fails to activate other associated downstream molecules involved in delayed apoptosis of host. Also, a significant reduction in cIAP levels was observed. Under oxidative exposure, inhibition of PERK phosphorylation debilitates infected RAW cell’s ability to maintain redox homeostasis leading to higher nitric oxide production. Altogether, L.donovani infection modulates host apoptosis in a PERK dependent manner and favours infection.
Collapse
Affiliation(s)
- Kumar Abhishek
- Division of Molecular Biology, Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, Bihar, India
| | - Sushmita Das
- Department of Microbiology, All India Institute of Medical Sciences, Phulwarisharif, Patna Bihar, India
| | - Ashish Kumar
- Division of Molecular Biology, Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, Bihar, India
| | - Ajay Kumar
- Division of Molecular Biology, Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, Bihar, India
| | - Vinod Kumar
- Division of Molecular Biology, Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, Bihar, India
| | - Savita Saini
- National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park, Hajipur, Vaishali, Bihar, India
| | - Abhishek Mandal
- Division of Molecular Biology, Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, Bihar, India
| | - Sudha Verma
- Division of Molecular Biology, Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, Bihar, India
| | - Manjay Kumar
- Division of Molecular Biology, Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, Bihar, India
| | - Pradeep Das
- Division of Molecular Biology, Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, Bihar, India
- * E-mail:
| |
Collapse
|
45
|
Solano-Gálvez SG, Abadi-Chiriti J, Gutiérrez-Velez L, Rodríguez-Puente E, Konstat-Korzenny E, Álvarez-Hernández DA, Franyuti-Kelly G, Gutiérrez-Kobeh L, Vázquez-López R. Apoptosis: Activation and Inhibition in Health and Disease. Med Sci (Basel) 2018; 6:E54. [PMID: 29973578 PMCID: PMC6163961 DOI: 10.3390/medsci6030054] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 06/28/2018] [Accepted: 06/29/2018] [Indexed: 12/16/2022] Open
Abstract
There are many types of cell death, each involving multiple and complex molecular events. Cell death can occur accidentally when exposed to extreme physical, chemical, or mechanical conditions, or it can also be regulated, which involves a genetically coded complex machinery to carry out the process. Apoptosis is an example of the latter. Apoptotic cell death can be triggered through different intracellular signalling pathways that lead to morphological changes and eventually cell death. This is a normal and biological process carried out during maturation, remodelling, growth, and development in tissues. To maintain tissue homeostasis, regulatory, and inhibitory mechanisms must control apoptosis. Paradoxically, these same pathways are utilized during infection by distinct intracellular microorganisms to evade recognition by the immune system and therefore survive, reproduce and develop. In cancer, neoplastic cells inhibit apoptosis, thus allowing their survival and increasing their capability to invade different tissues and organs. The purpose of this work is to review the generalities of the molecular mechanisms and signalling pathways involved in apoptosis induction and inhibition. Additionally, we compile the current evidence of apoptosis modulation during cancer and Leishmania infection as a model of apoptosis regulation by an intracellular microorganism.
Collapse
Affiliation(s)
- Sandra Georgina Solano-Gálvez
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico.
| | - Jack Abadi-Chiriti
- Departamento de Microbiología, Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte, Huixquilucán Estado de México 52786, México.
| | - Luis Gutiérrez-Velez
- Departamento de Microbiología, Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte, Huixquilucán Estado de México 52786, México.
| | - Eduardo Rodríguez-Puente
- Departamento de Microbiología, Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte, Huixquilucán Estado de México 52786, México.
| | - Enrique Konstat-Korzenny
- Departamento de Microbiología, Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte, Huixquilucán Estado de México 52786, México.
| | - Diego-Abelardo Álvarez-Hernández
- Departamento de Microbiología, Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte, Huixquilucán Estado de México 52786, México.
| | - Giorgio Franyuti-Kelly
- Medical IMPACT, Infectious Disease Department, Mexico City 53900, Estado de México, Mexico.
| | - Laila Gutiérrez-Kobeh
- Unidad de Investigación UNAM-INC, División Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México, Instituto Nacional de Cardiología, Mexico City, 14080, Mexico.
| | - Rosalino Vázquez-López
- Departamento de Microbiología, Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte, Huixquilucán Estado de México 52786, México.
| |
Collapse
|
46
|
Aslani F, Sebastian T, Keidel M, Fröhlich S, Elsässer HP, Schuppe HC, Klug J, Mahavadi P, Fijak M, Bergmann M, Meinhardt A, Bhushan S. Resistance to apoptosis and autophagy leads to enhanced survival in Sertoli cells. Mol Hum Reprod 2018; 23:370-380. [PMID: 28379541 DOI: 10.1093/molehr/gax022] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 03/31/2017] [Indexed: 12/13/2022] Open
Abstract
STUDY QUESTION What is the underlying mechanism of Sertoli cell (SC) resistance to cell death? SUMMARY ANSWER High expression of prosurvival B-cell lymphoma-2 (BCL2) proteins and inhibition of apoptosis and autophagy prolongs SC survival upon exposure to stress stimuli. WHAT IS KNOWN ALREADY In human and in experimental models of orchitis, tolerogenic SC survive stress conditions, while germ cells undergo massive apoptosis. In general, non-dividing highly differentiated cells tend to resist stress conditions for a longer time by favoring activation of prosurvival mechanisms and inhibition of cell death pathways. STUDY DESIGN, SIZE, DURATION In this cross sectional study, conditions stimulating apoptosis and autophagy were used to induce cell death in primary rat SC. Primary rat peritubular cells (PTC) and immortalized rat 93RS2 SC were used as controls. Each cell isolation was counted as one experiment (n = 1), and each experiment was repeated three to six times. PARTICIPANTS/MATERIALS, SETTING, METHODS Testis biopsy samples from infertile or subfertile patients and testis samples from rats with experimental autoimmune orchitis were used for immunohistological analysis. Primary SC were isolated from 19-day-old male Wistar rats. To maintain cell purity, cells were cultured in serum-free medium for apoptosis experiments and in medium supplemented with 1% serum for autophagy analyses. To induce apoptosis, cells were stimulated with staurosporine, borrelidin, cisplatin and etoposide for 4 or 24 h. Caspase three activation was examined by immunoblotting and enzymatic activity assay. Mitochondrial membrane potential was measured using tetramethylrhodamine methyl ester followed by flow cytometric analysis. Cytochrome c release was monitored by immunofluorescence. Cell viability was determined using the methylthiazole tetrazolium assay. To monitor autophagy flux, cells were deprived of nutrients using Hank's balanced salt solution for 1, 2 and 3 h. Formation of autophagosomes was analyzed by using immunoblotting, immunofluorescence labeling and ultrastructural analyses. Relative mRNA levels of genes involved in the regulation of apoptosis and autophagy were evaluated. Extracellular high mobility group box protein one was measured as a marker of necrosis using ELISA. MAIN RESULTS AND THE ROLE OF CHANCE SC survive the inflammatory conditions in vivo in human testis and in experimental autoimmune orchitis. Treatment with apoptosis inducing chemotherapeutics did not cause caspase three activation in isolated rat SC. Moreover, mitochondrial membrane potential and mitochondrial localization of cytochrome c were not changed by treatment with staurosporine, suggesting a premitochondrial blockade of apoptosis in SC. Expression levels of prosurvival BCL2 family members were significantly higher in SC compared to PTC at both mRNA and protein levels. Furthermore, after nutrient starvation, autophagy signaling was initiated in SC as observed by decreased levels of phosphorylated UNC- 51-like kinase -1 (ULK1). However, levels of light chain 3 II (LC3 II) and sequestosome1 (SQSTM1) remained unchanged, indicating blockade of the autophagy flux. Lysosomal activity was intact in SC as shown by accumulation of LC3 II following administration of lysosomal protease inhibitors, indicating that inhibition of autophagy flux occurs at a preceding stage. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION In this study, we have used primary SC from prepubertal rats. Caution should be taken when translating our results to adult animals, where crosstalk with other testicular cells and hormonal factors may also play a role in regulating survival of SC. WIDER IMPLICATIONS OF THE FINDINGS Our results suggest that inhibition of autophagy and apoptosis following exposure to extrinsic stress stimuli promotes SC survival, and is a possible mechanism to explain the robustness of SC in response to stress. Cell death resistance in SC is crucial for the recovery of spermatogenesis after chemotherapy treatment in cancer patients. Additionally, understanding the molecular mechanisms of SC survival unravels valuable target proteins, such as BCL2, that may be manipulated therapeutically to control cell viability depending on the context of the disease. STUDY FUNDING AND COMPETING INTEREST(S) This study was funded by the Deutsche Forschungsgemeinschaft (DFG) Grant BH93/1-1, and by the International Research Training Group between Justus Liebig University of Giessen and Monash University, Melbourne (GRK 1871/1) funded by the DFG and Monash University. The support of the Medical Faculty of Justus-Liebig University of Giessen is gratefully acknowledged. The authors declare no conflict of interest.
Collapse
Affiliation(s)
- Ferial Aslani
- Department of Anatomy and Cell Biology, Justus-Liebig University, Aulweg 123, Giessen 35392, Germany
| | - Tim Sebastian
- Department of Anatomy and Cell Biology, Justus-Liebig University, Aulweg 123, Giessen 35392, Germany
| | - Miguel Keidel
- Department of Anatomy and Cell Biology, Justus-Liebig University, Aulweg 123, Giessen 35392, Germany
| | - Suada Fröhlich
- Department of Anatomy and Cell Biology, Justus-Liebig University, Aulweg 123, Giessen 35392, Germany
| | - Hans-Peter Elsässer
- Department of Cell Biology and Cytopathology, Philipps University of Marburg, Germany
| | - Hans-Christian Schuppe
- Department of Urology, Pediatric Urology and Andrology, Justus-Liebig University, Giessen, Germany
| | - Jörg Klug
- Department of Anatomy and Cell Biology, Justus-Liebig University, Aulweg 123, Giessen 35392, Germany
| | - Poornima Mahavadi
- Department of Internal Medicine, Justus-Liebig University, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Monika Fijak
- Department of Anatomy and Cell Biology, Justus-Liebig University, Aulweg 123, Giessen 35392, Germany
| | - Martin Bergmann
- Institute of Veterinary Anatomy, Histology, and Embryology, Justus-Liebig-University, Giessen, Germany
| | - Andreas Meinhardt
- Department of Anatomy and Cell Biology, Justus-Liebig University, Aulweg 123, Giessen 35392, Germany
| | - Sudhanshu Bhushan
- Department of Anatomy and Cell Biology, Justus-Liebig University, Aulweg 123, Giessen 35392, Germany
| |
Collapse
|
47
|
Rodríguez-González J, Wilkins-Rodríguez AA, Gutiérrez-Kobeh L. Role of glutathione, ROS, and Bcl-xL in the inhibition of apoptosis of monocyte-derived dendritic cells by Leishmania mexicana promastigotes. Parasitol Res 2018; 117:1225-1235. [PMID: 29476339 DOI: 10.1007/s00436-018-5804-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 02/08/2018] [Indexed: 12/31/2022]
Abstract
Dendritic cells (DCs) are one of the principal host cells of the obligate intracellular parasite Leishmania that can survive and reproduce within cells due to the ability to regulate different cellular events, including apoptosis. Inhibition of host cell apoptosis is a strategy employed by multiple pathogens to ensure their survival in the infected cell. We have previously reported that Leishmania mexicana promastigotes and amastigotes inhibit camptothecin-induced apoptosis of monocyte-derived dendritic cells (moDCs) through the downregulation of p38 and JNK phosphorylation. The upregulation of glutathione (GSH), the most important regulator of reactive oxygen species (ROS) concentration, has proven to protect cells from apoptosis through the inhibition of JNK1. Another mechanism employed by cells for the protection of apoptosis is the expression of anti-apoptotic proteins of the Bcl-2 family. The aim of this study was to determine if GSH, ROS, and Bcl-xL participate in the inhibition of camptothecin-induced apoptosis of moDC by L. mexicana promastigotes. GSH quantification assays showed that camptothecin and BSO (an inhibitor of glutathione synthesis) strongly decreased intracellular GSH concentration in moDC, while infection with L. mexicana promastigotes had no effect in the level of GSH. On the other hand, infection with L. mexicana promastigotes of BSO- and camptothecin-treated moDC diminished the concentration of ROS and induced the expression of the anti-apoptotic protein Bcl-xL. Our findings suggest that inhibition of camptothecin-induced apoptosis of moDC by L. mexicana promastigotes is preferentially regulated by the expression of anti-apoptotic proteins of the Bcl-2 family rather than by the redox status of the cell.
Collapse
Affiliation(s)
- Jorge Rodríguez-González
- Unidad Periférica de Investigación UNAM-INC, División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México-Instituto Nacional de Cardiología "Ignacio Chávez", Juan Badiano No. 1, Col. Belisario Domínguez, Sección XVI, Delegación Tlalpan, C.P, 14080, Ciudad de México, México.,Posgrado en Ciencias Biológicas, Facultad de Medicina, Unidad de Posgrado, Ciudad Universitaria, 04510, Ciudad de México, México
| | - Arturo A Wilkins-Rodríguez
- Unidad Periférica de Investigación UNAM-INC, División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México-Instituto Nacional de Cardiología "Ignacio Chávez", Juan Badiano No. 1, Col. Belisario Domínguez, Sección XVI, Delegación Tlalpan, C.P, 14080, Ciudad de México, México
| | - Laila Gutiérrez-Kobeh
- Unidad Periférica de Investigación UNAM-INC, División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México-Instituto Nacional de Cardiología "Ignacio Chávez", Juan Badiano No. 1, Col. Belisario Domínguez, Sección XVI, Delegación Tlalpan, C.P, 14080, Ciudad de México, México.
| |
Collapse
|
48
|
Vishwakarma P, Parmar N, Chandrakar P, Sharma T, Kathuria M, Agnihotri PK, Siddiqi MI, Mitra K, Kar S. Ammonium trichloro [1,2-ethanediolato-O,O']-tellurate cures experimental visceral leishmaniasis by redox modulation of Leishmania donovani trypanothione reductase and inhibiting host integrin linked PI3K/Akt pathway. Cell Mol Life Sci 2018; 75:563-588. [PMID: 28900667 PMCID: PMC11105478 DOI: 10.1007/s00018-017-2653-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 08/11/2017] [Accepted: 09/05/2017] [Indexed: 10/18/2022]
Abstract
In an endeavor to search for affordable and safer therapeutics against debilitating visceral leishmaniasis, we examined antileishmanial potential of ammonium trichloro [1,2-ethanediolato-O,O']-tellurate (AS101); a tellurium based non toxic immunomodulator. AS101 showed significant in vitro efficacy against both Leishmania donovani promastigotes and amastigotes at sub-micromolar concentrations. AS101 could also completely eliminate organ parasite load from L. donovani infected Balb/c mice along with significant efficacy against infected hamsters (˃93% inhibition). Analyzing mechanistic details revealed that the double edged AS101 could directly induce apoptosis in promastigotes along with indirectly activating host by reversing T-cell anergy to protective Th1 mode, increased ROS generation and anti-leishmanial IgG production. AS101 could inhibit IL-10/STAT3 pathway in L. donovani infected macrophages via blocking α4β7 integrin dependent PI3K/Akt signaling and activate host MAPKs and NF-κB for Th1 response. In silico docking and biochemical assays revealed AS101's affinity to form thiol bond with cysteine residues of trypanothione reductase in Leishmania promastigotes leading to its inactivation and inducing ROS-mediated apoptosis of the parasite via increased Ca2+ level, loss of ATP and mitochondrial membrane potential along with metacaspase activation. Our findings provide the first evidence for the mechanism of action of AS101 with excellent safety profile and suggest its promising therapeutic potential against experimental visceral leishmaniasis.
Collapse
Affiliation(s)
- Preeti Vishwakarma
- Division of Parasitology, CSIR-Central Drug Research Institute, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh, 226031, India
- Academy of Scientific and Innovative Research, Anusandhan Bhawan, New Delhi, India
| | - Naveen Parmar
- Division of Parasitology, CSIR-Central Drug Research Institute, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh, 226031, India
- Academy of Scientific and Innovative Research, Anusandhan Bhawan, New Delhi, India
| | - Pragya Chandrakar
- Division of Parasitology, CSIR-Central Drug Research Institute, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh, 226031, India
- Academy of Scientific and Innovative Research, Anusandhan Bhawan, New Delhi, India
| | - Tanuj Sharma
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Manoj Kathuria
- Electron Microscopy Unit, Sophisticated Analytical Instrument Facility, CSIR-Central Drug Research Institute, Lucknow, India
| | - Pramod K Agnihotri
- Division of Toxicology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Mohammad Imran Siddiqi
- Academy of Scientific and Innovative Research, Anusandhan Bhawan, New Delhi, India
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Kalyan Mitra
- Academy of Scientific and Innovative Research, Anusandhan Bhawan, New Delhi, India
- Electron Microscopy Unit, Sophisticated Analytical Instrument Facility, CSIR-Central Drug Research Institute, Lucknow, India
| | - Susanta Kar
- Division of Parasitology, CSIR-Central Drug Research Institute, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh, 226031, India.
- Academy of Scientific and Innovative Research, Anusandhan Bhawan, New Delhi, India.
| |
Collapse
|
49
|
Immunomodulation of dual specificity phosphatase 4 during visceral leishmaniasis. Microbes Infect 2018; 20:111-121. [DOI: 10.1016/j.micinf.2017.10.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 10/30/2017] [Accepted: 10/31/2017] [Indexed: 02/07/2023]
|
50
|
Thomas SA, Nandan D, Kass J, Reiner NE. Countervailing, time-dependent effects on host autophagy promotes intracellular survival of Leishmania. J Biol Chem 2017; 293:2617-2630. [PMID: 29269416 DOI: 10.1074/jbc.m117.808675] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 12/18/2017] [Indexed: 11/06/2022] Open
Abstract
Autophagy is essential for cell survival under stress and has also been implicated in host defense. Here, we investigated the interactions between Leishmania donovani, the main etiological agent of visceral leishmaniasis, and the autophagic machinery of human macrophages. Our results revealed that during early infection-and via activation of the Akt pathway-Leishmania actively inhibits the induction of autophagy. However, by 24 h, Leishmania switched from being an inhibitor to an overall inducer of autophagy. These findings of a dynamic, biphasic response were based on the accumulation of lipidated light chain 3 (LC3), an autophagosome marker, by Western blotting and confocal fluorescence microscopy. We also present evidence that Leishmania induces delayed host cell autophagy via a mechanism independent of reduced activity of the mechanistic target of rapamycin (mTOR). Notably, Leishmania actively inhibited mTOR-regulated autophagy even at later stages of infection, whereas there was a clear induction of autophagy via some other mechanism. In this context, we examined host inositol monophosphatase (IMPase), reduced levels of which have been implicated in mTOR-independent autophagy, and we found that IMPase activity is significantly decreased in infected cells. These findings indicate that Leishmania uses an alternative pathway to mTOR to induce autophagy in host macrophages. Finally, RNAi-mediated down-regulation of host autophagy protein 5 (ATG5) or autophagy protein 9A (ATG9A) decreased parasite loads, demonstrating that autophagy is essential for Leishmania survival. We conclude that Leishmania uses an alternative pathway to induce host autophagy while simultaneously inhibiting mTOR-regulated autophagy to fine-tune the timing and magnitude of this process and to optimize parasite survival.
Collapse
Affiliation(s)
- Sneha A Thomas
- From the Department of Medicine, Division of Infectious Diseases, University of British Columbia, Vancouver, British Columbia V6H 3Z6 and
| | - Devki Nandan
- From the Department of Medicine, Division of Infectious Diseases, University of British Columbia, Vancouver, British Columbia V6H 3Z6 and
| | - Jennifer Kass
- From the Department of Medicine, Division of Infectious Diseases, University of British Columbia, Vancouver, British Columbia V6H 3Z6 and
| | - Neil E Reiner
- From the Department of Medicine, Division of Infectious Diseases, University of British Columbia, Vancouver, British Columbia V6H 3Z6 and .,the Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| |
Collapse
|